1
|
Liang J, Yang F, Li Z, Li Q. Epigenetic regulation of the inflammatory response in stroke. Neural Regen Res 2025; 20:3045-3062. [PMID: 39589183 PMCID: PMC11881735 DOI: 10.4103/nrr.nrr-d-24-00672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/15/2024] [Accepted: 09/20/2024] [Indexed: 11/27/2024] Open
Abstract
Stroke is classified as ischemic or hemorrhagic, and there are few effective treatments for either type. Immunologic mechanisms play a critical role in secondary brain injury following a stroke, which manifests as cytokine release, blood-brain barrier disruption, neuronal cell death, and ultimately behavioral impairment. Suppressing the inflammatory response has been shown to mitigate this cascade of events in experimental stroke models. However, in clinical trials of anti-inflammatory agents, long-term immunosuppression has not demonstrated significant clinical benefits for patients. This may be attributable to the dichotomous roles of inflammation in both tissue injury and repair, as well as the complex pathophysiologic inflammatory processes in stroke. Inhibiting acute harmful inflammatory responses or inducing a phenotypic shift from a pro-inflammatory to an anti-inflammatory state at specific time points after a stroke are alternative and promising therapeutic strategies. Identifying agents that can modulate inflammation requires a detailed understanding of the inflammatory processes of stroke. Furthermore, epigenetic reprogramming plays a crucial role in modulating post-stroke inflammation and can potentially be exploited for stroke management. In this review, we summarize current findings on the epigenetic regulation of the inflammatory response in stroke, focusing on key signaling pathways including nuclear factor-kappa B, Janus kinase/signal transducer and activator of transcription, and mitogen-activated protein kinase as well as inflammasome activation. We also discuss promising molecular targets for stroke treatment. The evidence to date indicates that therapeutic targeting of the epigenetic regulation of inflammation can shift the balance from inflammation-induced tissue injury to repair following stroke, leading to improved post-stroke outcomes.
Collapse
Affiliation(s)
- Jingyi Liang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| | - Qian Li
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Senapati S, Bertolini TB, Minnier MA, Yazicioglu MN, Markusic DM, Zhang R, Wicks J, Nahvi A, Herzog RW, Walsh MC, Cejas PJ, Armour SM. Inhibition of IFNAR-JAK signaling enhances tolerability and transgene expression of systemic non-viral DNA delivery. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102502. [PMID: 40206655 PMCID: PMC11979999 DOI: 10.1016/j.omtn.2025.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Lipid nanoparticles (LNPs) have demonstrated significant therapeutic value for non-viral delivery of mRNA and siRNA. While there is considerable interest in utilizing LNPs for delivering DNA (DNA-LNPs) to address a broad range of genetic disorders, acute inflammatory responses pose significant safety concerns and limit transgene expression below therapeutically relevant levels. However, the mechanisms and immune signaling pathways underlying DNA-LNP-triggered inflammatory responses are not well characterized. Through the use of gene-targeted mouse models, we have identified cGAS-STING and interferon-α/β receptor (IFNAR) pathways as major mediators of acute inflammation triggered by systemic delivery of DNA-LNPs. cGAS-STING activation induces expression of numerous JAK-STAT-activating cytokines, and we show that treatment of mice with the JAK inhibitors ruxolitinib or baricitinib significantly improves tolerability to systemically delivered DNA-LNPs. Furthermore, specific inhibition of IFNAR signaling enhances both DNA-LNP tolerability and transgene expression. Utilization of JAK inhibitors or IFNAR blockade represent promising strategies for enhancing the safety and efficacy of non-viral DNA delivery for gene therapy.
Collapse
Affiliation(s)
| | - Thais B. Bertolini
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - David M. Markusic
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rui Zhang
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Joan Wicks
- Gene Therapy Research, Spark Therapeutics, Philadelphia, PA, USA
| | - Ali Nahvi
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Roland W. Herzog
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Pedro J. Cejas
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Sean M. Armour
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| |
Collapse
|
3
|
Franzoni G, Signorelli F, Donniacuo A, Schiavo L, Napoletano M, De Matteis G, Grandoni F, Zinellu S, Bove V, Dei Giudici S, De Carlo E, Galiero G, Napolitano F, Martucciello A. Exploring potential cytokine profiles as diagnostic biomarkers for brucellosis in Mediterranean Buffaloes. Front Vet Sci 2025; 12:1583858. [PMID: 40406273 PMCID: PMC12097277 DOI: 10.3389/fvets.2025.1583858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/11/2025] [Indexed: 05/26/2025] Open
Abstract
Brucellosis is a zoonotic disease, with an important economic impact on the livestock industry and public health worldwide. Both Brucella abortus and Brucella melitensis can infect Mediterranean Buffalo (Bubalus bubalis), leading to infertility and abortion. In ruminants, the standard diagnostic approach involves two serological tests, the Rose Bengal Test and the Complement Fixation Test, applied in parallel, though their specificity requires improvement. Cytokines play a crucial role in coordinating immune responses through complex networks and can serve as biomarkers for various diseases. This study explored the potential use of cytokines as immunological biomarkers for Brucella infection in Mediterranean Buffalo. For this purpose, we included 18 healthy and 20 Brucella-infected buffaloes in our analysis. Heparinized blood samples were stimulated with the Brucella antigen, with PBS as nil control and PWM as lymphocyte viability control. After 16-24 h, plasma levels of IL-1α, IL-1β, IL-4, IL-6, IL-10, IL-17, IL-36Ra, MIP-1α, MIP-1β, MCP-1, CXCL8, IP-10, IFN-γ, TNF, and VEGF-A were measured using multiplex ELISA. Our results showed that infected animals released significantly higher levels of IFN-γ, IP-10, MCP-1 in response to Brucella antigen compared to healthy controls. Conversely, healthy animals released instead higher levels of IL-1α, IL-1β, IL-6 and IL-10 following antigen stimulation compared to infected animals. Finally, sequential canonical discriminant analyses were performed to generate predictive cytokine profiles for each group. The findings indicated that a combination of five cytokines (IFN-γ, IP-10, IL-1α, IL-1β, IL-6) can effectively distinguished infected from healthy buffaloes. Overall, this study suggests that incorporating these key immune cytokines could improve the diagnostic accuracy of brucellosis in Mediterranean Buffalo.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Federica Signorelli
- Council for Agricultural Research and Economics (CREA)- Research Centre for Animal Production and Aquaculture, Monterotondo (RM), Italy
| | - Anna Donniacuo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Lorena Schiavo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Michele Napoletano
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Giovanna De Matteis
- Council for Agricultural Research and Economics (CREA)- Research Centre for Animal Production and Aquaculture, Monterotondo (RM), Italy
| | - Francesco Grandoni
- Council for Agricultural Research and Economics (CREA)- Research Centre for Animal Production and Aquaculture, Monterotondo (RM), Italy
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Vincenzo Bove
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Esterina De Carlo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Giorgio Galiero
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Francesco Napolitano
- Council for Agricultural Research and Economics (CREA)- Research Centre for Animal Production and Aquaculture, Monterotondo (RM), Italy
| | - Alessandra Martucciello
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| |
Collapse
|
4
|
Heidari-Japelaghi R, Valizadeh M, Haddad R. Interferon gamma-induced hub genes and key pathways: A study based on biological network analysis and experimental validation. J Biotechnol 2025; 405:72-87. [PMID: 40348089 DOI: 10.1016/j.jbiotec.2025.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
By performing a biological network analysis, we identified some hub genes, which were up- or down-regulated in the breast cancer (BC) cell line after treatment with IFN-γ. Moreover, several pathways including cytokine-cytokine receptor interaction, TNF signaling pathway, NOD-like receptor signaling pathway, and NF-κB signaling pathway were detected that their activation leads to the antiproliferation, proapoptosis, and antiviral activities. To validate in silico results, the bioactivity of recombinant human IFN-γ (hIFN-γ) produced in different hosts was analyzed by antiviral and anticancer assays. The antiviral role of the hIFN-γ preparations was evaluated by inhibition of Vesicular Stomatitis Virus (VSV)-mediated cytopathic effects on Vero cells. A dose-dependent increase in cell viability was observed at different concentrations of recombinant proteins. The maximum amount of the cell viability detected for the hIFN-γ preparations was determined at a concentration of 32.00 pg/mL. To analyze the cytotoxic efficacy of the hIFN-γ preparations on the growth and development of tumor cells, a BC cell line (MCF-7) was treated with both recombinant protein forms in a time- and dose-dependent way. The highest level of inhibiting cell proliferation was detected at a concentration of 32.00 pg/mL hIFN-γ after 72 h incubation. Anticancer and antiviral functions of IFN-γ were confirmed via the expression analysis of hub genes cd74, cxcl10, il6, and stat1 using RT-PCR. Furthermore, the hIFN-γ preparations were significantly able to up-regulate the expression of proapoptotic Bax and p53 and to down-regulate Bcl-2 as an antiapoptotic gene, showing the cytotoxic effect of hIFN-γ toward MCF-7 cells via apoptosis induction.
Collapse
Affiliation(s)
- Reza Heidari-Japelaghi
- Department of Biotechnology Engineering, Faculty of Agriculture and Natural Resources, Imam Khomeini International University, Qazvin, Iran.
| | - Mostafa Valizadeh
- Department of Plant Breeding and Biotechnology, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Raheem Haddad
- Department of Biotechnology Engineering, Faculty of Agriculture and Natural Resources, Imam Khomeini International University, Qazvin, Iran
| |
Collapse
|
5
|
David S, Castro L, Duarte E, Gaspar U, da Costa Rodrigues MR, Cueto-Rojo MV, Mendonça J, Ferrão J, Machado M, Poças J, Lavinha J, Vieira L, Santos AS. Genetic variants in the IFNGR2 locus associated with severe chronic Q fever. Hum Immunol 2025; 86:111271. [PMID: 40056764 DOI: 10.1016/j.humimm.2025.111271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/27/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025]
Abstract
Q fever is a highly contagious zoonosis capable of causing large outbreaks of important health and economic consequences. Host genetic factors are believed to influence the development of severe chronic Q fever following the infection by the etiological agent, Coxiella burnetii. Targetted next generation sequencing (NGS) was performed in a case-control genetic association study on 53 confirmed Q fever cases, including 38 compatible with acute and 15 with chronic disease, and 29 samples from the general Portuguese population. Four SNPs in the IFNGR2 locus, rs78407108 G > A, rs17879956 C > T, rs7277167 C > T, and rs9974603 C > A, showed a statistically significant association to chronic Q fever, resisting the Bonferroni correction. These belonged to haplotypes significantly associated with chronic Q fever. The individual SNPs are referenced in the GTEx database as possible eQTLs. Given the direct bearing of IFNGR2 on IFN-γ signaling, the possible involvement of the associated variants with higher IFNGR2 expression could be in line with observations suggesting that IFN-γ production in chronic Q fever patients is significantly higher than in healthy controls. Further investigations are required to clarify the role of IFNGR2 signaling in association with chronic Q fever.
Collapse
Affiliation(s)
- Susana David
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal; Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmacia, Universidade de Lisboa, Portugal
| | - Liliana Castro
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal
| | - Elsa Duarte
- MED - Instituto Mediterrâneo para a Agricultura, AmbientePortugal e Desenvolvimento, Escola de Ciências e Tecnologia, Universidade de Evora, Portugal
| | - Ulisses Gaspar
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal
| | - Maria Rosário da Costa Rodrigues
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal; Patologia Clínica - Lab. Bioquímica Genética/Endocrinologia Especial, Hosp. D. Estefânia, Unidade Local de Saúde São José, Lisboa, Portugal; BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisboa, Portugal
| | - Maria Vanessa Cueto-Rojo
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal
| | - Joana Mendonça
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal
| | - José Ferrão
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal
| | - Miguel Machado
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal
| | - José Poças
- Serviço de Infeciologia, Hospital de São Bernardo, Centro Hospitalar de Setúbal, Setúbal, Portugal
| | - João Lavinha
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal
| | - Luís Vieira
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, I. P. (INSA), Lisboa, Portugal
| | - Ana Sofia Santos
- Centro de Estudos de Vectores e Doenças Infecciosas Dr. Francisco Cambournac (CEVDI), Departamento de Doenças Infeciosas (INSA), Águas de Moura, Portugal; Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
6
|
Wu M, Liu J, Liu L, Yang Y, Liu H, Yu L, Zeng H, Yuan S, Xu R, Liu H, Jiang H, Qu S, Wang L, Chen Y, Wang J, Zhang Y, He S, Feng L, Han J, Zeng W, Wang H, Huang Y. Autologous Peripheral Vγ9Vδ2 T Cell Synergizes with αβ T Cell Through Antigen Presentation and BTN3A1 Blockade in Immunotherapy of Cervical Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401230. [PMID: 40091603 PMCID: PMC12079532 DOI: 10.1002/advs.202401230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/07/2025] [Indexed: 03/19/2025]
Abstract
New treatment strategies are urgently needed for patients with advanced cervical cancer (CC). Here, a synergistic anti-CC effect of a novel combinatorial immunotherapy with adoptively transferred autologous Vγ9Vδ2 T cells and αβ T cells is shown. The pivotal role of both circulating and tumor-infiltrating Vγ9Vδ2 T cells in anti-CC immunity is uncovered. Importantly, autologous Vγ9Vδ2 T cells show a synergistic anti-CC effect with αβ T cells not only through killing tumor directly, but also by promoting the activation and tumoricidal activity of syngeneic αβ T cells through antigen presentation, which can be further boosted by conventional chemotherapy. Moreover, Vγ9Vδ2 T cells can restore the tumoricidal function of αβ T cell through competitively binding to BTN3A1, a TCR-Vγ9Vδ2 ligand on CC cells upregulated by IFN-γ derived from activated αβ T cell. These findings uncover a critical synergistic effect of autologous Vγ9Vδ2 T cells and αβ T cells in immunotherapy of CC and reveal the underlying mechanisms.
Collapse
Affiliation(s)
- Min Wu
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Jian Liu
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Liting Liu
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Yifan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hong Liu
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Long Yu
- Beckman Coulter Commercial Enterprise (China) Co., LtdShanghai200122China
| | - Haihong Zeng
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shuo Yuan
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Ruiyi Xu
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Hangyu Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Han Jiang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shen Qu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Liming Wang
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Jingyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Yuwei Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shan He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Junyan Han
- Department of Immunology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Wanjiang Zeng
- Department of Obstetrics and Gynecology, Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hui Wang
- Department of Obstetrics and GynecologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyCancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineZhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
7
|
Fu H, Sun W, Xu Y, Zhang H. Advances in cytokine gene polymorphisms in tuberculosis. mSphere 2025; 10:e0094424. [PMID: 40162798 PMCID: PMC12039272 DOI: 10.1128/msphere.00944-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Tuberculosis (TB), especially pulmonary tuberculosis (PTB), is a prevalent infectious disease affecting the respiratory system and is characterized by high morbidity, disability, and mortality rates that significantly impact the quality of life of patients and their families. Host genetic susceptibility plays a crucial role in the infection process of Mycobacterium tuberculosis (M. tuberculosis) with single nucleotide polymorphisms (SNPs) identified as key factors in the genetic loci associated with tuberculosis occurrence and progression. Research indicates that polymorphisms in cytokine genes-including interferons, interleukins, tumor necrosis factors, and chemokines-are closely linked to the onset, progression, and treatment outcomes of pulmonary tuberculosis. Investigating cytokine gene polymorphisms in PTB patients is essential for understanding disease mechanisms and prognosis. This review summarizes the role of cytokine polymorphisms in tuberculosis morbidity, elucidates the biological genetic mechanisms involved at the molecular level, and provides insights into clinical treatment strategies for TB.
Collapse
Affiliation(s)
- Haiyang Fu
- Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Jiangsu, China
| | - Wenqiang Sun
- Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Jiangsu, China
| | - Ye Xu
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Haiyun Zhang
- Department of Laboratory, Dalian Municipal Women and Children’s Medical Center, Dalian, Liaoning, China
| |
Collapse
|
8
|
Qi P, Jiang X, Wang X, Sheng L, Liang J, Zhang L. Unraveling the pathogenesis and prevention strategies of acute high-altitude illness through gut microecology. NPJ Biofilms Microbiomes 2025; 11:62. [PMID: 40263277 PMCID: PMC12015534 DOI: 10.1038/s41522-025-00701-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
High-altitude environments, characterized by hypobaric and hypoxic conditions, induce acute hypoxia, resulting in decreased blood oxygen saturation. This hypoxic stress perturbs gut microecological homeostasis, significantly contributing to the pathogenesis of acute mountain sickness. Consequently, elucidating the mechanisms by which high altitude affects gut homeostasis is crucial for developing effective interventions.
Collapse
Affiliation(s)
- Ping Qi
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, PR China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Precision Medicine Laboratory, the First Hospital of Lanzhou University, Lanzhou, 730000, PR China
| | - Xiansen Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, PR China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Precision Medicine Laboratory, the First Hospital of Lanzhou University, Lanzhou, 730000, PR China
| | - Xiaojuan Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, PR China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Precision Medicine Laboratory, the First Hospital of Lanzhou University, Lanzhou, 730000, PR China
| | - Liang Sheng
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, PR China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Precision Medicine Laboratory, the First Hospital of Lanzhou University, Lanzhou, 730000, PR China
| | - Jiawen Liang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, PR China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China
- Precision Medicine Laboratory, the First Hospital of Lanzhou University, Lanzhou, 730000, PR China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, PR China.
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China.
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China.
- Precision Medicine Laboratory, the First Hospital of Lanzhou University, Lanzhou, 730000, PR China.
- Clinical Research Center for General Surgery of Gansu Province, Lanzhou, 730000, PR China.
- Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou, 730000, PR China.
| |
Collapse
|
9
|
Westfall S, Gentile ME, Olsen TM, Karo-Atar D, Bogza A, Röstel F, Pardy RD, Mandato G, Fontes G, Herbert D, Melichar HJ, Abadie V, Richer MJ, Vinh DC, Koenig JFE, Harrison OJ, Divangahi M, Weis S, Gregorieff A, King IL. A type 1 immune-stromal cell network mediates disease tolerance against intestinal infection. Cell 2025:S0092-8674(25)00395-2. [PMID: 40267906 DOI: 10.1016/j.cell.2025.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 12/03/2024] [Accepted: 03/26/2025] [Indexed: 04/25/2025]
Abstract
Type 1 immunity mediates host defense through pathogen elimination, but whether this pathway also impacts tissue function is unknown. Here, we demonstrate that rapid induction of interferon γ (IFNγ) signaling coordinates a multicellular response that is critical to limit tissue damage and maintain gut motility following infection of mice with a tissue-invasive helminth. IFNγ production is initiated by antigen-independent activation of lamina propria CD8+ T cells following MyD88-dependent recognition of the microbiota during helminth-induced barrier invasion. IFNγ acted directly on intestinal stromal cells to recruit neutrophils that limited parasite-induced tissue injury. IFNγ sensing also limited the expansion of smooth muscle actin-expressing cells to prevent pathological gut dysmotility. Importantly, this tissue-protective response did not impact parasite burden, indicating that IFNγ supports a disease tolerance defense strategy. Our results have important implications for managing the pathophysiological sequelae of post-infectious gut dysfunction and chronic inflammatory diseases associated with stromal remodeling.
Collapse
Affiliation(s)
- Susan Westfall
- Department of Microbiology and Immunology, Department of Medicine, Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada; McGill Centre for Microbiome Research, Montreal, QC, Canada
| | - Maria E Gentile
- Department of Microbiology and Immunology, Department of Medicine, Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada; Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tayla M Olsen
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Danielle Karo-Atar
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Andrei Bogza
- Department of Microbiology and Immunology, Department of Medicine, Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada; McGill Centre for Microbiome Research, Montreal, QC, Canada
| | - Franziska Röstel
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany; Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Ryan D Pardy
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laval, QC, Canada
| | - Giordano Mandato
- Department of Microbiology and Immunology, Department of Medicine, Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada; McGill Centre for Microbiome Research, Montreal, QC, Canada
| | - Ghislaine Fontes
- Department of Microbiology and Immunology, Department of Medicine, Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada; McGill Centre for Microbiome Research, Montreal, QC, Canada
| | - De'Broski Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather J Melichar
- Department of Microbiology and Immunology, McGill University Montreal, Montreal, QC, Canada
| | - Valerie Abadie
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Martin J Richer
- Department of Microbiology and Immunology, McGill University Montreal, Montreal, QC, Canada; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Donald C Vinh
- Department of Microbiology and Immunology, Department of Medicine, Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Joshua F E Koenig
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Oliver J Harrison
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Maziar Divangahi
- Department of Microbiology and Immunology, Department of Medicine, Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Sebastian Weis
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany; Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany; Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Alex Gregorieff
- Department of Pathology, McGill University and Cancer Research Program, Research Institute of McGill University Health Centre, Montreal, QC, Canada; McGill Regenerative Medicine Network, Montreal, QC, Canada
| | - Irah L King
- Department of Microbiology and Immunology, Department of Medicine, Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada; McGill Centre for Microbiome Research, Montreal, QC, Canada; McGill Regenerative Medicine Network, Montreal, QC, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, QC, Canada.
| |
Collapse
|
10
|
Cuisiniere T, Hajjar R, Oliero M, Calvé A, Fragoso G, Rendos HV, Gerkins C, Taleb N, Gagnon-Konamna M, Dagbert F, Loungnarath R, Sebajang H, Schwenter F, Wassef R, Ratelle R, De Broux É, Richard C, Santos MM. Initial gut microbiota composition is a determining factor in the promotion of colorectal cancer by oral iron supplementation: evidence from a murine model. MICROBIOME 2025; 13:100. [PMID: 40259408 PMCID: PMC12013013 DOI: 10.1186/s40168-025-02101-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) development is influenced by both iron and gut microbiota composition. While iron supplementation is routinely used to manage anemia in CRC patients, it may also impact gut microbiota and promote tumorigenesis. In this study, we investigated the impact of initial gut microbiota composition on iron-promoted tumorigenesis. We performed fecal microbiota transplantation (FMT) in ApcMin/+ mice using samples from healthy controls, CRC patients, and mice, followed by exposure to iron sufficient or iron excess diets. RESULTS We found that iron supplementation promoted CRC and resulted in distinct gut microbiota changes in ApcMin/+ mice receiving FMT from CRC patients (FMT-CRC), but not from healthy controls or mice. Oral treatment with identified bacterial strains, namely Faecalibaculum rodentium, Holdemanella biformis, Bifidobacterium pseudolongum, and Alistipes inops, protected FMT-CRC mice against iron-promoted tumorigenesis. CONCLUSIONS Our findings suggest that microbiota-targeted interventions may mitigate tumorigenic effects of iron supplementation in anemic patients with CRC.
Collapse
Affiliation(s)
- Thibault Cuisiniere
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Roy Hajjar
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Manon Oliero
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Gabriela Fragoso
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Hervé Vennin Rendos
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Claire Gerkins
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Nassima Taleb
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Marianne Gagnon-Konamna
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - François Dagbert
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Rasmy Loungnarath
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Herawaty Sebajang
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Frank Schwenter
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Ramses Wassef
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Richard Ratelle
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Éric De Broux
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Carole Richard
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Manuela M Santos
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada.
- Institut du Cancer de Montréal, Montréal, Québec, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
11
|
Raballah E, Anyona SB, Osata SW, Wasena SA, Onyango C, Hurwitz I, Cheng Q, Seidenberg PD, McMahon BH, Ouma C, Ong'echa JM, Schneider KA, Perkins DJ. Impact of age, HIV1, sickle-cell genotypes, and interferon-gamma gene upstream variants on malaria disease outcomes in a longitudinal pediatric cohort. Sci Rep 2025; 15:13043. [PMID: 40234522 PMCID: PMC12000329 DOI: 10.1038/s41598-025-97267-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
This prospective cohort study explored the association between two upstream IFN-γ variants (rs2069709: G > T and rs2069705: A > G) and hazard factors for malaria outcomes in a longitudinal cohort of children (n = 941, 3-36 mos.), followed for three years. The impact of age, sex, previous malaria exposure, HIV1 infection, and sickle-cell genotypes (HbAA, HbAS, and HbSS) was also investigated. Reduced malaria episodes were associated with older age at enrollment [HR = 0.957 (95% CI = 0.953-0.961) per month, P < 2.2e-16], HIV1 infection [0.687 (0.545-0.866), P = 0.001], being female [0.910 (0.859-0.964), P = 0.040], and HbAS [0.823 (0.754-0.898), P = 0.005]. The GA/TA diplotype [0.376 (0.230-0.614), P = 0.002] also reduced the hazard of malaria, while TA haplotype increased susceptibility [1.749 (1.159-2.640), P = 0.029]. Factors protecting against the development of SMA [Hemoglobin (Hb < 6.0 g/dL)] included older age [0.927 (0.913-0.942) per month, P < 2.2e-16], previous malaria episodes [0.576 (0.542-0.614, P = 9.5e-32)], HbAS [0.553 (0.400-0.766), P = 0.015]. The rs2069705AG genotype increased the hazard of SMA [1.697 (1.002-2.875), P = 0.042]. Reduced hazard of mortality was observed for older children [0.898 (0.857-0.941), P < 2.2e-16], while a higher hazard was present in HIV-infected children [12.475 (6.380-24.392), P < 2.2e-16], and in those with HbSS [6.341 (1.944-20.686), P = 0.007]. The GG haplotype increased the mortality hazard [1.817 (0.936-3.527), P = 0.078]. The results here highlight critical factors influencing the hazard of malaria, SMA, and mortality.
Collapse
Affiliation(s)
- Evans Raballah
- Department of Medical Laboratory Sciences, School of Public Health Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology, Kakamega, Kenya
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
| | - Samuel B Anyona
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Medical Biochemistry, School of Medicine, Maseno University, Maseno, Kenya
| | - Shamim W Osata
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Sharley A Wasena
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Clinton Onyango
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Ivy Hurwitz
- Department of Internal Medicine, Center for Global Health, University of New Mexico, 913 Camino de Salud, IDTC 3140, Albuquerque, NM, 87131, USA
| | - Qiuying Cheng
- Department of Internal Medicine, Center for Global Health, University of New Mexico, 913 Camino de Salud, IDTC 3140, Albuquerque, NM, 87131, USA
| | - Philip D Seidenberg
- Department of Emergency Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Benjamin H McMahon
- Theoretical Division, Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Collins Ouma
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - John M Ong'echa
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Kristan A Schneider
- Department of Internal Medicine, Center for Global Health, University of New Mexico, 913 Camino de Salud, IDTC 3140, Albuquerque, NM, 87131, USA
| | - Douglas J Perkins
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya.
- Department of Internal Medicine, Center for Global Health, University of New Mexico, 913 Camino de Salud, IDTC 3140, Albuquerque, NM, 87131, USA.
| |
Collapse
|
12
|
Nygrén P, Bouhlal J, Jokinen E, Forstén S, Laajala E, Dias D, Adnan-Awad S, Ianevski A, Klievink J, Lähteenmäki H, Kuusanmäki H, Myllymäki M, Kasanen T, Saeed K, Lee DA, iCAN Study Group, Hjorth-Hansen H, Aittokallio T, Dufva O, Mustjoki S. High-throughput drug screening identifies SMAC mimetics as enhancers of NK-cell cytotoxicity in chronic myeloid leukemia. Blood 2025; 145:1670-1686. [PMID: 39792962 PMCID: PMC12000656 DOI: 10.1182/blood.2024025286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/31/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Natural killer (NK) cells have proven to be safe and effective immunotherapies, associated with favorable treatment responses in chronic myeloid leukemia (CML). Augmenting NK-cell function with oncological drugs could improve NK-cell-based immunotherapies. Here, we used a high-throughput drug screen consisting of >500 small-molecule compounds, to systematically evaluate the effects of oncological drugs on primary NK cells against CML cells. We identified second mitochondrially derived activator of caspases (SMAC) mimetics as potent enhancers of NK-cell cytotoxicity in both cell lines and primary patient samples. In contrast, several drug classes, including glucocorticoids and tyrosine kinase inhibitors such as dasatinib, inhibited NK-cell cytotoxicity. Single-cell RNA sequencing revealed drug-induced transcriptomic changes in both NK and target CML cells. SMAC mimetics upregulated NF-κB target genes in NK cells, potentially contributing to their enhanced cytotoxicity. Inhibitory drugs dexamethasone, dasatinib, and sotrastaurin prevented NK-cell transition to an activated state and suppressed the expression of interferon gamma (IFN-γ) by NK cells, thus preventing IFN-γ-mediated target cell transcriptomic response. In conclusion, we discovered that SMAC mimetics sensitize cancer cells to NK-cell-mediated killing, with potential clinical applications especially in patients with advanced phase CML.
Collapse
MESH Headings
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/drug effects
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- High-Throughput Screening Assays
- Cytotoxicity, Immunologic/drug effects
- Antineoplastic Agents/pharmacology
- Mitochondrial Proteins
- Apoptosis Regulatory Proteins
- Cell Line, Tumor
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Petra Nygrén
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Jonas Bouhlal
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Emmi Jokinen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Sofia Forstén
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Essi Laajala
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Diogo Dias
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Shady Adnan-Awad
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Aleksandr Ianevski
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Jay Klievink
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Hanna Lähteenmäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Mikko Myllymäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Tiina Kasanen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Khalid Saeed
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Dean A. Lee
- Division of Hematology, Oncology, and Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, OH
| | - iCAN Study Group
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Division of Hematology, Oncology, and Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, OH
- Department of Hematology, St. Olavs Hospital, Trondheim, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | | | - Tero Aittokallio
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Olli Dufva
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Chen Y, Chen R, Li H, Shuai Z. Clinical management of autoimmune liver diseases: juncture, opportunities, and challenges ahead. Immunol Res 2025; 73:67. [PMID: 40195209 PMCID: PMC11976385 DOI: 10.1007/s12026-025-09622-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
The three major autoimmune liver diseases are autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), and primary sclerosing cholangitis (PSC).These conditions are assumed to result from a breakdown in immunological tolerance, which leads to an inflammatory process that causes liver damage.The self-attack is started by T-helper cell-mediated identification of liver autoantigens and B-cell production of autoantibodies,and it is maintained by a reduction in the number and activity of regulatory T-cells.Infections and environmental factors have been explored as triggering factors for these conditions, in addition to a genetic predisposition.Allelic mutations in the HLA locus have been linked to vulnerability, as have relationships with single nucleotide polymorphisms in non-HLA genes.Despite the advances in the management of these diseases, there is no curative treatment for these disorders, and a significant number of patients eventually progress to an end-stage liver disease requiring liver transplantation.In this line, tailored immune-therapeutics have emerged as possible treatments to control the disease.In addition, early diagnosis and treatment are pivotal for reducing the long-lasting effects of these conditions and their burden on quality of life.Herein we present a review of the etiology, clinical presentation, diagnosis, and challenges on ALDs and the feasible solutions for these complex diseases.
Collapse
MESH Headings
- Humans
- Hepatitis, Autoimmune/therapy
- Hepatitis, Autoimmune/diagnosis
- Hepatitis, Autoimmune/immunology
- Hepatitis, Autoimmune/etiology
- Cholangitis, Sclerosing/therapy
- Cholangitis, Sclerosing/diagnosis
- Cholangitis, Sclerosing/immunology
- Liver Cirrhosis, Biliary/therapy
- Liver Cirrhosis, Biliary/diagnosis
- Liver Cirrhosis, Biliary/immunology
- Animals
- Immunotherapy/methods
- Autoimmune Diseases/therapy
- Autoimmune Diseases/diagnosis
- Disease Management
- Genetic Predisposition to Disease
Collapse
Affiliation(s)
- Yangfan Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Ruofei Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Haiyan Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zongwen Shuai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, China.
| |
Collapse
|
14
|
Vandendriessche K, van Suylen V, Brouckaert J, Matthys P, Dauwe D, Meyns B, Erasmus M, Neyrinck A, Rex S, Rega F. The role of a hemoadsorption filter on cytokine levels during 1 hour of thoraco-abdominal normothermic regional perfusion for donation after circulatory death heart donation in a porcine model. Artif Organs 2025; 49:615-626. [PMID: 39665498 DOI: 10.1111/aor.14924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/26/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Both global ischemia caused by circulatory arrest and extracorporeal circulation circuits have been shown to trigger cytokine release. We hypothesized that inserting a hemoadsorption device during thoraco-abdominal normothermic regional perfusion (TA-NRP) in the donation after circulatory death setting would mitigate the inflammatory response, potentially resulting in improved cardiac allograft function. METHODS In 15 pigs, circulatory arrest was induced by hypoxia. After a 15-min no-touch-period, TA-NRP was performed for 60 min. Eight pigs had a hemoadsorption device incorporated in the ECC, while seven did not. Plasma concentrations of IFN-α, IFN-γ, TNF-α, IL-1β, IL-4, IL-6, IL-8, IL-10, and IL-12p40 were assessed by ELISA at baseline, immediately at start of TA-NRP, 60 min after start of TA-NRP (just before weaning from ECC), and at 30 and 60 min after weaning from ECC. Cardiac function was assessed with pressure-volume loop analysis. RESULTS Hemoadsorption had no relevant effects on systemic cytokine levels post TA-NRP. IL-6 plasma levels gradually rose throughout the procedure for both groups. Hemoadsorption did not affect systolic or diastolic left ventricular function, nor were global hemodynamics improved by hemoadsorption. CONCLUSIONS The insertion of a hemoadsorption device did not significantly affect plasma cytokine levels or cardiac function. Further research is necessary to assess the role of the inflammatory response in DCD heart transplantation and its modulation by TA-NRP.
Collapse
Affiliation(s)
| | - Vincent van Suylen
- Department of Cardiothoracic Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Janne Brouckaert
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Dieter Dauwe
- Department of Intensive Care, University Hospitals Leuven, Leuven, Belgium
| | - Bart Meyns
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Michiel Erasmus
- Department of Cardiothoracic Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arne Neyrinck
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, Catholic University Leuven, Leuven, Belgium
| | - Steffen Rex
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Filip Rega
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Lin Y, Yang L, Li Y, Dou S, Zhang Z, Zhou Q. CD4+CD25- T-Cell-Secreted IFN-γ Promotes Corneal Nerve Degeneration in Diabetic Mice. Invest Ophthalmol Vis Sci 2025; 66:15. [PMID: 40192636 PMCID: PMC11980951 DOI: 10.1167/iovs.66.4.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Purpose This study aimed to explore the relationship between corneal nerve degeneration and elevated dendritic cells (DCs) in diabetic keratopathy. Methods Corneas from diabetic and healthy mice were analyzed using single-cell RNA sequencing. Corneal nerve density and DC and T-cell infiltration were quantified through whole-mount corneal staining. Freshly isolated mouse trigeminal ganglion (TG) neurons were co-cultured with immature DCs, mature DCs, activated CD8+ T cells, and CD4+CD25- T cells. TG neurite outgrowth was assessed to identify potential effector cells driving corneal nerve degeneration. In addition, interferon-gamma (IFN-γ) and blocking antibodies were used to evaluate their effects on TG neurite outgrowth and corneal nerve degeneration in mice. Results Compared with age-matched healthy mice, diabetic mice exhibited a significant reduction in corneal nerve density and sensitivity, along with increased infiltration of DCs, CD4+ T cells, and CD8+ T cells. In vitro co-culture experiments revealed that CD4+CD25- T cells, rather than DCs and CD8+ T cells, significantly inhibited TG neurite outgrowth. Among cytokines, elevated IFN-γ in diabetic corneas impaired TG neurite outgrowth and induced corneal nerve degeneration, whereas IL-4 and IL-17 had no such effect. Blocking IFN-γ alleviated CD4+CD25- T-cell-induced inhibition of TG neurite outgrowth and corneal nerve degeneration in diabetic mice. Conclusions CD4+CD25- T cells, but not DCs or CD8+ T cells, contribute to corneal nerve degeneration in diabetic mice, a process partially mediated by IFN-γ.
Collapse
Affiliation(s)
- Yujing Lin
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Lingling Yang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Ya Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Zhenzhen Zhang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| |
Collapse
|
16
|
Smith TA, Zhou L, Ghergherehchi CL, Mikesh M, Yang CZ, Tucker HO, Allgood J, Bushman JS, Bittner GD. Polyethylene glycol has immunoprotective effects on sciatic allografts, but behavioral recovery and graft tolerance require neurorrhaphy and axonal fusion. Neural Regen Res 2025; 20:1192-1206. [PMID: 38989956 PMCID: PMC11438327 DOI: 10.4103/nrr.nrr-d-23-01220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/26/2023] [Accepted: 02/29/2024] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00033/figure1/v/2024-07-06T104127Z/r/image-tiff Behavioral recovery using (viable) peripheral nerve allografts to repair ablation-type (segmental-loss) peripheral nerve injuries is delayed or poor due to slow and inaccurate axonal regeneration. Furthermore, such peripheral nerve allografts undergo immunological rejection by the host immune system. In contrast, peripheral nerve injuries repaired by polyethylene glycol fusion of peripheral nerve allografts exhibit excellent behavioral recovery within weeks, reduced immune responses, and many axons do not undergo Wallerian degeneration. The relative contribution of neurorrhaphy and polyethylene glycol-fusion of axons versus the effects of polyethylene glycol per se was unknown prior to this study. We hypothesized that polyethylene glycol might have some immune-protective effects, but polyethylene glycol-fusion was necessary to prevent Wallerian degeneration and functional/behavioral recovery. We examined how polyethylene glycol solutions per se affect functional and behavioral recovery and peripheral nerve allograft morphological and immunological responses in the absence of polyethylene glycol-induced axonal fusion. Ablation-type sciatic nerve injuries in outbred Sprague-Dawley rats were repaired according to a modified protocol using the same solutions as polyethylene glycol-fused peripheral nerve allografts, but peripheral nerve allografts were loose-sutured (loose-sutured polyethylene glycol) with an intentional gap of 1-2 mm to prevent fusion by polyethylene glycol of peripheral nerve allograft axons with host axons. Similar to negative control peripheral nerve allografts not treated by polyethylene glycol and in contrast to polyethylene glycol-fused peripheral nerve allografts, animals with loose-sutured polyethylene glycol peripheral nerve allografts exhibited Wallerian degeneration for all axons and myelin degeneration by 7 days postoperatively and did not recover sciatic-mediated behavioral functions by 42 days postoperatively. Other morphological signs of rejection, such as collapsed Schwann cell basal lamina tubes, were absent in polyethylene glycol-fused peripheral nerve allografts but commonly observed in negative control and loose-sutured polyethylene glycol peripheral nerve allografts at 21 days postoperatively. Loose-sutured polyethylene glycol peripheral nerve allografts had more pro-inflammatory and less anti-inflammatory macrophages than negative control peripheral nerve allografts. While T cell counts were similarly high in loose-sutured-polyethylene glycol and negative control peripheral nerve allografts, loose-sutured polyethylene glycol peripheral nerve allografts expressed some cytokines/chemokines important for T cell activation at much lower levels at 14 days postoperatively. MHCI expression was elevated in loose-sutured polyethylene glycol peripheral nerve allografts, but MHCII expression was modestly lower compared to negative control at 21 days postoperatively. We conclude that, while polyethylene glycol per se reduces some immune responses of peripheral nerve allografts, successful polyethylene glycol-fusion repair of some axons is necessary to prevent Wallerian degeneration of those axons and immune rejection of peripheral nerve allografts, and produce recovery of sensory/motor functions and voluntary behaviors. Translation of polyethylene glycol-fusion technologies would produce a paradigm shift from the current clinical practice of waiting days to months to repair ablation peripheral nerve injuries.
Collapse
Affiliation(s)
- Tyler A. Smith
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Liwen Zhou
- Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| | | | - Michelle Mikesh
- Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Cathy Z. Yang
- Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Haley O. Tucker
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - JuliAnne Allgood
- Division of Pharmaceutical Sciences, University of Wyoming, Laramie, WY, USA
| | - Jared S. Bushman
- Division of Pharmaceutical Sciences, University of Wyoming, Laramie, WY, USA
| | - George D. Bittner
- Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
17
|
Libramento ZP, Tichy L, Parry TL. Muscle wasting in cancer cachexia: Mechanisms and the role of exercise. Exp Physiol 2025. [PMID: 40159295 DOI: 10.1113/ep092544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025]
Abstract
Cancer cachexia (CC) is a multifactorial disease marked by a severe and progressive loss of lean muscle mass and characterized further by inflammation and a negative energy/protein balance, ultimately leading to muscle atrophy and loss of muscle tissue. As a result, patients experiencing cachexia have reduced muscle function and thus less independence and a lower quality of life. CC progresses through stages of increasing severity: pre-cachexia, cachexia and refractory cachexia. Two proposed underlying mechanisms that drive cancer-induced muscle wasting are the autophagy-lysosome and ubiquitin-proteasome systems. An increase in autophagic flux and proteolytic activity leads to atrophy of both cardiac and skeletal muscle, ultimately mediated by tumour or immune-secreted inflammatory cytokines. These pathways occur at a basal level to maintain cellular homeostasis; therefore, it is the overactivation of the pathways that leads to muscle atrophy. Recent evidence demonstrates the ability of aerobic and resistance training to restore these pathways to their basal levels. The mechanism is not yet understood, and more research is needed to determine exactly how exercise influences each pathway. However, exercise has great promise as a therapeutic strategy for CC because of the evidence for it preserving muscle mass and function, and attenuating protein degradative pathways. The extent to which exercise affects the ubiquitin-proteasome and autophagy-lysosome systems is determined by the frequency, intensity and duration of the exercise protocol. As such, an ideal exercise prescription is lacking for individuals with CC.
Collapse
Affiliation(s)
- Zoe P Libramento
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, North Carolina, USA
| | - Louisa Tichy
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, North Carolina, USA
| | - Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, North Carolina, USA
| |
Collapse
|
18
|
Al-Janabi A, Martin P, Simpson C, Rhys H, Khan AR, Eyre S, Christofi M, Foulkes AC, Skelton A, Viatte S, Barton A, Morris AP, Smith CH, Griffiths CEM, Warren RB. Blood Single-Cell Transcriptomic and Proteomic Signatures of Paradoxical Eczema in Patients with Psoriasis Treated with Biologics. J Invest Dermatol 2025:S0022-202X(25)00370-7. [PMID: 40157420 DOI: 10.1016/j.jid.2025.02.153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/10/2025] [Accepted: 02/07/2025] [Indexed: 04/01/2025]
Abstract
Biologics targeting the TNF and IL-17/23 axis are highly effective treatments for psoriasis but can result in cutaneous adverse events. The pathogenesis of paradoxical eczema, the occurrence of an atopic dermatitis phenotype after biologic initiation in people with psoriasis, is unknown. Using single-cell RNA sequencing and mass cytometry, we found increased expression of TNF, IFN-γ, and IFN-α and their signaling pathways in paradoxical eczema case cell clusters compared with that in matched psoriasis controls. Genetic variants influencing the expression of chemokine signaling and TNF pathway genes were associated with paradoxical eczema in a separate genotyped cohort, and this association was independent of known atopic risk loci. This suggests that paradoxical eczema has a predominantly type 1 systemic inflammatory signature and that genetic susceptibility to aberrant chemokine and TNF pathway signaling could contribute to development of this phenotype during biologic treatment.
Collapse
Affiliation(s)
- Ali Al-Janabi
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom.
| | - Paul Martin
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | | | | | | | - Steve Eyre
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom; Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom
| | - Maria Christofi
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom
| | - Amy C Foulkes
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | | | - Sebastien Viatte
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Catherine H Smith
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom; St. John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Christopher E M Griffiths
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom; St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom; Department of Dermatology, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Richard B Warren
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
19
|
Jooss T, Maier K, Reichardt LM, Hindelang B, Süberkrüb L, Hamberger KL, Bülow JM, Schuetze K, Gebhard F, Mannes M, Halbgebauer R, Wohlgemuth L, Huber-Lang M, Relja B, Bergmann CB. Dynamic functional assessment of T cells reveals an early suppression correlating with adverse outcome in polytraumatized patients. Front Immunol 2025; 16:1538516. [PMID: 40196124 PMCID: PMC11973370 DOI: 10.3389/fimmu.2025.1538516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Most trauma patients require intensive care treatment and are susceptible to developing persistent inflammation and immunosuppression, potentially leading to multi organ dysfunction syndrome (MODS) and dependence on long term care facilities. T cells undergo changes in numbers and function post trauma. T cell dysfunction in polytraumatized patients was characterized using functional immunomonitoring to predict individual clinical outcome. Moreover, the potential to reverse T cell dysfunction using Interleukin (IL)-7 was examined. Methods Blood samples were drawn from healthy individuals and prospectively enrolled polytrauma patients (Injury Severity Score ≥ 18) on admission, 8, 24 and 48 hours, 5 and 10 days after. CD3/28-stimulated cytokine production of T cells in whole blood was assessed via Enzyme Linked Immuno Spot (ELISpot). T cell subsets were quantified via counting and flow cytometry. Unfavorable physical performative outcome was defined as death or new functional disability necessitating long term care. Secondary outcomes were the development of MODS and in-hospital mortality. IL-7 was added ex vivo to test reversibility of cytokine disturbances. Results 34 patients were enrolled. The different outcome groups showed no difference in injury severity. Patients with favorable physical performative outcome revealed higher functional T cell specific Interferon γ (IFN-γ) and IL-17 (8 hours) and lower IL-10 production (day 5) and higher CD8 T cell concentrations. Patients without MODS development showed a higher IFN-γ (day 10), higher IL-2 (8 hours) and higher IL-17 production (admission, day 5). There were no differences regarding in-hospital mortality. Systemic blood IFN-γ, IL-2 and IL-10 concentrations only correlated with MODS (24 hours). Systemic CD8 T cell numbers correlated with functional IFN-γ production. Whole blood stimulation with IL-7 increased functional T cell IFN-γ release. Discussion Our study reveals an early characteristic overall T cell dysfunction of pro-inflammatory (IFN-γ, IL-2, IL-17) and immunosuppressive (IL-10) subtypes in polytraumatized patients. Our data indicates that rather the functional capacity of T cells to release cytokines, but not systemic cytokine concentrations can be used to predict outcome post trauma. We assume that the early stimulation of pro- and anti-inflammatory T cells benefits polytraumatized patients. Potentiation of functional IFN-γ release might be achieved by IL-7 administration.
Collapse
Affiliation(s)
- Tobias Jooss
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Katharina Maier
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Lena-Marie Reichardt
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Bianca Hindelang
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Lönna Süberkrüb
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Kim Lena Hamberger
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Jasmin Maria Bülow
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Konrad Schuetze
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Florian Gebhard
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Borna Relja
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Christian B. Bergmann
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
20
|
Ye X, Hsu CY, Jia L, Zhang X, Magee C, Whitham S, Leigh S, Evans JD, Zhang L, Robinson K. Dynamic immune response to Avian Pathogenic Escherichia coli infection in broiler chickens: Insights into pro-inflammatory and anti-inflammatory cytokine regulation: CYTOKINE REGULATION IN APEC INFECTED BROILERS. Poult Sci 2025; 104:105029. [PMID: 40220487 PMCID: PMC12018555 DOI: 10.1016/j.psj.2025.105029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 04/14/2025] Open
Abstract
Avian Pathogenic Escherichia coli (APEC) poses a significant threat to the U.S. poultry industry, causing respiratory infections and systemic colibacillosis. Understanding APEC's impact on the immune response is crucial for developing effective prevention and treatment strategies. This study investigates the dynamic immune responses to APEC infection in broiler chickens, with a focus on survival rates, airsac lesion scores, and cytokine gene expression patterns in lung tissue. Seven-day-old broiler chicks were divided into control and APEC-inoculated groups, with the control group receiving tryptic soy broth and the APEC group receiving 7 × 10^7 CFU of APEC via intratracheal inoculation. Survival data included both male and female birds, while airsac lesion score and lung tissue samples for gene expression analyses were collected only from male birds at nine time points post-infection (days 1, 3, 5, 7, 9, 11, 13, 15, and 21). High airsac lesion incidence and mortality were observed during early infection stages, decreasing in mid to later stages as anti-inflammatory cytokines were upregulated. The lung gene expression study analyzed the expression of pro-inflammatory cytokines (IFN-γ, IL-1β, IL-8, and IL-6), regulator (SOCS3), and anti-inflammatory cytokines (IL-10, TGFβ-2, TGFβ-3, and IL-1RN) via RT-qPCR assays, using 18S rRNA for normalization. A two-way ANOVA followed by Tukey's Kramer test evaluated the effects of APEC treatment and days post-infection on gene expression, with the Mann-Whitney U test comparing fold changes between groups. Results indicated an early upregulation of pro-inflammatory cytokines like IFN-γ and IL-1β, followed by the modulatory roles of SOCS3, TGF-β, and IL-1RN, balancing the immune response and possibly preventing excessive tissue damage. This study elucidates the dynamic regulation of key cytokines during APEC infection in chickens, providing insights into immune mechanisms. Understanding these mechanisms is crucial for developing targeted therapies and improving disease management in poultry, potentially reducing antibiotic dependence and enhancing overall poultry health and productivity.
Collapse
Affiliation(s)
- Xin Ye
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| | - Chuan-Yu Hsu
- Institute for Genomic, Biocomputing, and Biotechnology, Mississippi State University, Mississippi State, MS 39762, USA
| | - Linan Jia
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| | - Xue Zhang
- Department of Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Christopher Magee
- Poultry Research Unit, Agriculture Research Service, United States Department of Agriculture (USDA), Mississippi State, MS 39762, USA
| | - Stephanie Whitham
- Poultry Research Unit, Agriculture Research Service, United States Department of Agriculture (USDA), Mississippi State, MS 39762, USA
| | - Spencer Leigh
- Poultry Research Unit, Agriculture Research Service, United States Department of Agriculture (USDA), Mississippi State, MS 39762, USA
| | - Jeffrey D Evans
- Poultry Research Unit, Agriculture Research Service, United States Department of Agriculture (USDA), Mississippi State, MS 39762, USA
| | - Li Zhang
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| | - Kelsy Robinson
- Poultry Research Unit, Agriculture Research Service, United States Department of Agriculture (USDA), Mississippi State, MS 39762, USA.
| |
Collapse
|
21
|
Lin MH, Maniam P, Li D, Tang B, Bishop CR, Suhrbier A, Earl LW, Tayyar Y, McMillan NA, Li L, Harrich D. Harnessing defective interfering particles and lipid nanoparticles for effective delivery of an anti-dengue virus RNA therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102424. [PMID: 39817192 PMCID: PMC11733052 DOI: 10.1016/j.omtn.2024.102424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Currently, no approved antiviral drugs target dengue virus (DENV) infection, leaving treatment reliant on supportive care. DENV vaccine efficacy varies depending on the vaccine type, the circulating serotype, and vaccine coverage. We investigated defective interfering particles (DIPs) and lipid nanoparticles (LNPs) to deliver DI290, an anti-DENV DI RNA. Both DIPs and DI290-loaded LNPs (LNP-290) effectively suppressed DENV infection in human primary monocyte-derived macrophages (MDMs), THP-1 macrophages, and fibroblasts-natural DENV targets. Inhibiting interferon (IFN) signaling with a Janus kinase 1/2 inhibitor or an IFN-α/β receptor 1 (IFNAR1)-binding antibody blocked DIP and LNP-290 antiviral activity. LNP-290 demonstrated a greater than log10 inhibition of DENV viral loads in IFNAR-deficient (Ifnar -/- ) and IFN regulatory factor (IRF) 3 and 7 double knockout (Irf3/7 -/- ) mice. Pathway analysis of RNA sequencing data from LNP-treated C57BL/6J mice, Ifnar -/- mice, and human MDMs treated with LNPs or DENV DIPs indicated DI290 treatment enhanced IFN responses, suggesting IFN-λ and IFN-γ provided antiviral activity when IFN-α/β responses were diminished. While viral interference by DI290 is possible, results did not support RNA replication competition as an inhibition mechanism. These findings suggest that DI290 may be a promising DENV therapeutic by activating the innate immune system.
Collapse
Affiliation(s)
- Min-Hsuan Lin
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Pramila Maniam
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Dongsheng Li
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Bing Tang
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Cameron R. Bishop
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Andreas Suhrbier
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- Global Virus Network (GVN) Center of Excellence, Australian Infectious Disease Research Centre, Brisbane, QLD 4072, Australia
| | - Lucy Wales- Earl
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Yaman Tayyar
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
- Prorenata Biotech, Molendinar, QLD 4214, Australia
| | - Nigel A.J. McMillan
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD 4072, Australia
| | - David Harrich
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- Global Virus Network (GVN) Center of Excellence, Australian Infectious Disease Research Centre, Brisbane, QLD 4072, Australia
| |
Collapse
|
22
|
Leventelis C, Veskoukis AS, Rojas Gil AP, Papadopoulos P, Garderi M, Angeli A, Kampitsi A, Tsironi M. Methadone and Buprenorphine as Medication for Addiction Treatment Diversely Affect Inflammation and Craving Depending on Their Doses. PHARMACY 2025; 13:40. [PMID: 40126313 PMCID: PMC11932288 DOI: 10.3390/pharmacy13020040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/31/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Buprenorphine and methadone are widely used as medication for addiction treatment (MAT) in patients with opioid use disorders. However, there is no compelling evidence of their impact on the immune-endocrine response. Therefore, the aim of this study was to examine the effects of the aforementioned medications on craving and on biomarkers of inflammation and cortisol, approaching the dose issue concurrently. Sixty-six patients (thirty-four under methadone and thirty-two under buprenorphine) who had just entered a MAT program and were stabilized with the suitable administered doses after a two-week process were divided into four groups based on medication dose (i.e., methadone high dose, buprenorphine high dose, methadone medium dose, and buprenorphine medium dose). The heroin craving questionnaire for craving assessment was completed, and the blood biomarkers were measured on Days 1 and 180. According to the results, high doses of both medications were accompanied by low levels of craving, cortisol, and inflammation on Day 1, and no alterations were observed on Day 180. On the contrary, medium doses reduced the tested psychosocial and biochemical parameters in terms of time, indicating a positive action for the patients. Concludingly, modifications in MAT doses are needed soon after the stabilization process to prevent inflammation and avoid relapse, thus helping opioid-addicted patients toward rehabilitation.
Collapse
Affiliation(s)
- Christonikos Leventelis
- Department of Nursing, University of Peloponnese, 22100 Tripoli, Greece; (A.P.R.G.); (M.T.)
- Organization Against Drugs, 10433 Athens, Greece; (P.P.); (M.G.); (A.A.)
| | - Aristidis S. Veskoukis
- Department of Nutrition and Dietetics, University of Thessaly, Argonafton 1, 42132 Trikala, Greece;
| | - Andrea Paola Rojas Gil
- Department of Nursing, University of Peloponnese, 22100 Tripoli, Greece; (A.P.R.G.); (M.T.)
| | | | - Maria Garderi
- Organization Against Drugs, 10433 Athens, Greece; (P.P.); (M.G.); (A.A.)
| | - Asimina Angeli
- Organization Against Drugs, 10433 Athens, Greece; (P.P.); (M.G.); (A.A.)
| | | | - Maria Tsironi
- Department of Nursing, University of Peloponnese, 22100 Tripoli, Greece; (A.P.R.G.); (M.T.)
| |
Collapse
|
23
|
Li X, Xu H, Hong R, Yang H, Xu L, Zheng G, Xie B. Frontline pemetrexed and cisplatin based-chemotherapy combined with NRT promoted the antitumor in a mouse model of lung carcinoma. Int Immunopharmacol 2025; 149:114174. [PMID: 39929101 DOI: 10.1016/j.intimp.2025.114174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
The efficacy of neoantigen-reactive T cells (NRT) therapy in solid tumors, encompassing aspects such as infiltration, recognition, cytotoxicity, and enduring persistence, is notably influenced by the immunological microenvironment. This study endeavors to investigate whether the co-administration of pemetrexed and cisplatin augments the therapeutic efficacy of NRT therapy in lung cancer. Neoantigens were predicted using a comprehensive analysis of mutation data from Lewis lung carcinoma cells and mouse tail tissues. The immunogenicity of NRT cells was assessed through flow cytometry and IFN-γ ELISpot assays. A mouse model of NSCLC was used to investigate the anti-tumor effects of NRT combined with chemotherapy. The combination of NRT cells and chemotherapy significantly inhibited tumor growth in a mouse model, increased CD3+/CD137+ T cells to promote IFN-γ secretion from NRT cells, and up-regulated the levels of inflammatory cytokine proteins including IFN-γ, TNF, IL-6 and IL-10. Immunofluorescence analysis confirmed increased T-cell infiltration in tumor tissues without adverse effects on vital organs. In addition, transcriptome analyses indicated that the tumor microenvironment was altered to favor M1-like macrophages with an increased M1/M2 ratio, creating a pro-inflammatory environment. The integration of NRT with frontline chemotherapy for lung cancer could yield profoundly ideal therapeutic outcomes.
Collapse
Affiliation(s)
- Xiaoqin Li
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Hang Xu
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Rujun Hong
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Haitao Yang
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Lihuan Xu
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Guanying Zheng
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China.
| | - Baosong Xie
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China.
| |
Collapse
|
24
|
Agas D, Sabbieti MG. Untangling Ariadne's Thread Within the Bone Marrow Maze: A Close-Up View of Stem/Progenitor Cells' Interactome and Secretome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40035957 DOI: 10.1007/5584_2024_847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
The bone marrow (BM) is a multifactorial, highly dynamic, still not fully "mapped," reservoir. The BM labyrinthine landscape is subject to a relentless debate on the specialized and stem/progenitor cells' scattering within designated microareas. Certainly, BM tissue plays a watchdog role in bone modeling and remodeling, hematopoiesis, immune surveillance, and endocrine response integration. Parameters like tissue topographical distinctiveness, stiffness and porosity grade, and cells' behavioral idiosyncrasies in terms of stem/progenitor cell housing, activation, and motility represent a knotty problem not easily solved. Given that the disruption of BM microdomains has been associated with a number of severe pathological disorders, the comprehension and preservation of the BM workspace at multiple levels have become mandatory. Solid evidence has showed the existence of an intricate and tightly regulated cross-talk between the BM cellular occupants. Direct physical cell-cell connections and soluble mediators, including cytokines, chemokines, growth factors, exosomes and microvesicles, orchestrate composite intracellular signaling routes. The spatiotemporal action of definite biofactors ensures a functional blood-producing organ with a physiological bone turnover and prompts the action of multipotent stromal/hematopoietic cells. Recently, significant research efforts have been addressed to build bioengineered niche-mimic models based on biofunctionalized scaffolds and organoid-like constructs. These artificial BM niches combine and transduce various aspects of bioinformatics and tissue engineering to unravel the complexities of BM organization. This chapter aims to unfold the recent breakthroughs in the understanding of a BM intramural cell-cell dialogue in a physiological and, in some cases, within an inflammatory background. BM maze is gradually being discovered, but there is still a long way to go.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| | | |
Collapse
|
25
|
Kim Y, Hur J, Hong SC, Jung J, Park CH, Park JB, Yoon TJ, Kim JB, Yang SH. Modulated electro-hyperthermia therapy combined with Korean mistletoe extract treatment exerts a strong anti-tumor activity by enhancing cellular and humoral immune responses in mice. Anim Cells Syst (Seoul) 2025; 29:163-172. [PMID: 40040867 PMCID: PMC11878165 DOI: 10.1080/19768354.2025.2470455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
Electro-hyperthermia therapy (EHT) has been known to cause temperature-dependent cell death and enhance the effects of conventional antitumor treatments, such as chemotherapy and radiotherapy. Furthermore, EHT modulates the innate and adaptive immune systems. Mistletoe is one of the most broadly studied complementary and alternative therapeutic agents for cancer treatment due to its ability to stimulate the immune systems. This study aimed to investigate the effects of EHT and mistletoe therapy combination on immune responses. Tumors induced by B16-BL6 melanoma cells were treated twice with modulated EHT (mEHT) (43°C for 10 or 20 min) and with intravenous injection of a Korean mistletoe extract (KME). We examined the level of interferon (IFN)-γ, granzyme, interleukin (IL)-2, IL-10, and tumor-specific antibodies using enzyme-linked immunosorbent assay methods to further study the immunological responses in the combination of mEHT and KME. Additionally, cytotoxic T lymphocyte (CTL) activity is investigated. In this study, we revealed a significant anti-tumor immunological activity elevation in tumor-bearing mice by combined mEHT and KME therapy. Specifically, the combination of mEHT and KME treatment was effective in inhibiting tumor growth in mice. The combination treatment elicited CTL immune response and increased IFN-γ and granzyme secretion. Particularly, the co-treatment appeared to efficiently suppress the immune signal related to tumor-associated macrophage differentiation. Importantly, tumor cell-specific antibodies could be induced in mice after mEHT-treated tumor cell immunization, which represent a promising cancer vaccine strategy. Thus, our results indicate the therapeutic actions of KME as a feasible partner of mEHT, suggesting its potential candidate for cancer immunotherapy. Abbreviations: APC, Antigen-presenting cell; CTL, Cytotoxic T lymphocyte; EHT, Electro-hyperthermia therapy; ELISA, Enzyme-linked immunosorbent assay; HSP, Heat shock protein; KME, Korean mistletoe extract; NK, Natural killer; PBS, Phosphate-buffered saline; QOL, Quality of life; RF, Radio-frequency; TAM, Tumor-associated macrophage.
Collapse
Affiliation(s)
- Yebeen Kim
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, Republic of Korea
| | - Jinwoo Hur
- Department of Food and Nutrition, Yuhan University, Buchoen, Republic of Korea
| | - Sung-Chul Hong
- Department of Food Science and Biotechnology, Kunsan National University, Kunsan, Republic of Korea
| | - Jaewoon Jung
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, Republic of Korea
| | - Choon-Ho Park
- Graduate School of Clinical Pharmacy and Pharmaceutics, Ajou University, Suwon, South Korea
| | - Joon Beom Park
- Mistletoe Research Center, New Breath Hospital, Seoul, Republic of Korea
| | | | - Jong Bae Kim
- Mistletoe Research Center, New Breath Hospital, Seoul, Republic of Korea
| | - Seung-Hoon Yang
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Zhao W, Su J, Xue Q, Gao J, Bai H, Gao Y, Chen X, Liu W, Liu D, Wang G, Zhou X. Impact of foot-and-mouth disease virus on memory T and B cell populations in swine. Vet Microbiol 2025; 302:110406. [PMID: 39978867 DOI: 10.1016/j.vetmic.2025.110406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/22/2025]
Abstract
Foot-and-mouth disease virus (FMDV) is a highly contagious picornavirus that poses a serious threat to the global livestock industry. This study aimed to investigate the impact of FMDV infection on the memory immune response in pigs and to analyze the role of type II interferon (IFN-γ) in this process. By comparing pigs artificially infected with FMDV and those vaccinated with inactivated FMDV vaccine, we found that FMDV infection significantly suppressed the development of memory T helper (Th) and B cell populations, affecting the memory immune response. Further experiments showed that pretreatment with IFN-γ could counteract the immunosuppression caused by FMDV, and this counteraction was achieved by promoting the expression of three transcription factors: T-bet, Eomes, and Bcl-6. Our findings emphasize the key role of IFN-γ in regulating the host's immune response to FMDV infection and provide new scientific evidence for the development of effective FMDV vaccines.
Collapse
Affiliation(s)
- Wei Zhao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Jia Su
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Jie Gao
- Beijing Zhonghai Biotech Co., Ltd., Beijing 100081, China
| | - Hongxu Bai
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Yueyi Gao
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Xiaochun Chen
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Weijie Liu
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Dongdong Liu
- Inner Mongolia Bigvet Biotech Co., Ltd., Inner Mongolia 011500, China
| | - Guohua Wang
- Inner Mongolia Bigvet Biotech Co., Ltd., Inner Mongolia 011500, China
| | - Xiangmei Zhou
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
27
|
Ahmed B, Aliyu M, Getso MI, Bala JA, Ahmed RJ, Kabuga AI, Adamu AMY, Yusuf AA. Exploring the impact of interferon-gamma single nucleotide polymorphisms on HTLV-1 infection: Unraveling genetic influences in viral pathogenesis. Crit Rev Oncol Hematol 2025; 207:104614. [PMID: 39798937 DOI: 10.1016/j.critrevonc.2025.104614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/26/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025] Open
Abstract
Human T-lymphotropic virus-1 (HTLV-1) induces neoplastic adult T-cell leukemia/lymphoma (ATLL) and neurological HTLV-1 associated myelopathy (HAM) in approximately 3 %-5 % of infected individuals. The precise factors that facilitate disease manifestation are still unknown; interaction between the virus and the host's immune response is key. Cytokines regulates physiological activities and their dysregulation may initiate the pathogenesis of various malignant and infectious diseases. Genetic variations, particularly polymorphisms in gene regulatory regions, lead to varying cytokine production patterns. Interferon-gamma (IFN-γ), a key cytokine in HTLV-1 infection, is a signature cytokine for T-helper 1 (Th1) cells that interferes with viral replication and enhances innate and adaptive immune responses during viral infections. The IFNG gene possesses several single nucleotide polymorphisms (SNPs), among which the + 874 A/T SNP has been widely studied for its functional role in HTLV-1 infection. The purpose of this review was to provide insight into the impact of IFNG SNPs on HTLV-1 Infection.
Collapse
Affiliation(s)
- Bilkisu Ahmed
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria
| | - Mansur Aliyu
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria.
| | - Muhammad Ibrahim Getso
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria
| | - Jamilu Abubakar Bala
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria; Centre for Infectious Diseases Research, Bayero University Kano, P.M.B 3011, Kano, Nigeria
| | - Ramat Jummai Ahmed
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Ahmadu Bello University Zaria, P.M.B 1044, Zaria, Kaduna State, Nigeria
| | - Auwal Idris Kabuga
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria
| | - Al-Muktar Yahuza Adamu
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria
| | - Aminu Abba Yusuf
- Department of Haematology, Bayero University Kano and Aminu Kano Teaching Hospital, Kano, Nigeria
| |
Collapse
|
28
|
Song Y, Cui Y, Zhong Y, Wang Y, Zheng X. Fecal microbiota transplantation combined with inulin promotes the development and function of early immune organs in chicks. J Biotechnol 2025; 399:81-90. [PMID: 39826698 DOI: 10.1016/j.jbiotec.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Modern management of chicks hinders the vertical transmission of intestinal microbiota, which is closely related to immunity. Inulin is a substrate that can be utilized by the microbiota. This study aimed to determine whether fecal microbiota transplantation (FMT) combined with inulin played a "1 + 1 > 2" role in enhancing the development and function of immune organs. Chicks were treated with 1 % inulin and/or fecal microbiota suspension on days 1-6. The growth performance, immune organ development, and immune indicators were evaluated on days 7, 14, and 21. Results showed that the combination of FMT and inulin significantly increased the immune organ index on day 7 and promoted the morphological structure and the expression of proliferating cell nuclear antigen (PCNA) in immune organs on days 7, 14, and 21. Each treatment increased the gene expression of interferon-gamma (IFN-γ), interleukin-4 (IL-4), interleukin-2 (IL-2), B cell-activating factor receptor (BAFFR), B cell linker (BLNK), C-X-C Motif Chemokine Ligand 12 (CXCL12), C-X-C Motif Chemokine Receptor 4 (CXCR4), and Biotin (Bu-1) to varying degrees. FMT combined with inulin significantly increased the expression of IgA-positive cells on days 7 and 14. In conclusion, the synergistic effect of FMT and inulin had beneficial impacts on the development and function of immune organs.
Collapse
Affiliation(s)
- Yang Song
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Road, Nanguan District, Changchun, Jilin 130118, China
| | - Yibo Cui
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Road, Nanguan District, Changchun, Jilin 130118, China
| | - Yue Zhong
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Road, Nanguan District, Changchun, Jilin 130118, China
| | - Yumeng Wang
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Road, Nanguan District, Changchun, Jilin 130118, China
| | - Xin Zheng
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Road, Nanguan District, Changchun, Jilin 130118, China.
| |
Collapse
|
29
|
Maciel-Fiuza MF, Sbruzzi RC, Feira MF, Costa PDSS, Bonamigo RR, Vettorato R, Eidt LM, de Moraes PC, Oliveira Fam BSD, Castro SMDJ, Silveira MIDS, Vianna FSL. Influence of Cytokine-Related genetic variants in TNF, IL6, IL1β, and IFNγ genes in the thalidomide treatment for Erythema nodosum leprosum in a Brazilian population sample. Hum Immunol 2025; 86:111260. [PMID: 39956090 DOI: 10.1016/j.humimm.2025.111260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
Erythema nodosum leprosum (ENL), an inflammatory reaction in leprosy, causes painful nodules, fever, and malaise due to immune system activation. Thalidomide is an effective treatment, although associated with important adverse effects. We aimed to evaluate the association of genetic variants in genes encoding tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β) and interleukin-6 (IL-6) with the response to treatment of ENL with thalidomide. 148 patients from the South and Northeast regions of Brazil were included. Genomic DNA was isolated from blood and/or saliva samples using commercial kits, and genetic variants in TNF, IL6, IL1β, and IFNγ genes were genotyped by TaqMan system. We identified an association between polymorphisms in TNF (rs1799964C, rs1800630A, rs1799724T and rs1800629A) IL1β (rs4848306G, rs1143623G, rs16944A, and rs1143627A), IL6 (rs2069840C and rs2069845G) and IFNγ (rs2430561T) with thalidomide dose variation in a time-dependent manner. Associations of IL6 and TNF haplotypes with thalidomide dosage variation over the time of treatment were also observed. Polymorphisms in TNF, IL6, IL1β, and IFNγ genes may modulate their expression levels, potentially impacting the required dosage of thalidomide in the treatment of ENL. Our findings should be confirmed in further studies to estimate the size effect of these polymorphisms on ENL treatment with thalidomide.
Collapse
Affiliation(s)
- Miriãn Ferrão Maciel-Fiuza
- Postgraduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil; National Institute of Population Medical Genetics Porto Alegre Rio Grande do Sul Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre Porto Alegre Rio Grande do Sul Brazil; Immunobiology and Immunogenetics Laboratory, Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil
| | - Renan Cesar Sbruzzi
- Postgraduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre Porto Alegre Rio Grande do Sul Brazil; Immunobiology and Immunogenetics Laboratory, Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil
| | - Mariléa Furtado Feira
- Postgraduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil; National Institute of Population Medical Genetics Porto Alegre Rio Grande do Sul Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre Porto Alegre Rio Grande do Sul Brazil; Immunobiology and Immunogenetics Laboratory, Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil
| | | | - Renan Rangel Bonamigo
- Postgraduate Program in Pathology, Federal University of Health Sciences of Porto Alegre Porto Alegre Rio Grande do Sul Brazil; Dermatology Service of Hospital Santa Casa de Porto Alegre Porto Alegre Rio Grande do Sul Brazil; Postgraduate Program in Medicine, Medical Sciences, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil; Dermatology Service of Hospital de Clínicas de Porto Alegre Rio Grande do Sul Brazil
| | - Rodrigo Vettorato
- Dermatology Service of Hospital Santa Casa de Porto Alegre Porto Alegre Rio Grande do Sul Brazil
| | - Letícia Maria Eidt
- Sanitary Dermatology Outpatient Clinic, State Health Department of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil
| | - Paulo Cezar de Moraes
- Postgraduate Program in Medicine, Medical Sciences, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil; Sanitary Dermatology Outpatient Clinic, State Health Department of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil
| | - Bibiana Sampaio de Oliveira Fam
- Postgraduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil; National Institute of Population Medical Genetics Porto Alegre Rio Grande do Sul Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre Porto Alegre Rio Grande do Sul Brazil
| | - Stela Maris de Jezus Castro
- Department of Statistics, Universidade Federal Do Rio Grande Do Sul Porto Alegre Brazil; Postgraduate Program in Epidemiology, Universidade Federal Do Rio Grande Do Sul Porto Alegre Brazil
| | | | - Fernanda Sales Luiz Vianna
- Postgraduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil; National Institute of Population Medical Genetics Porto Alegre Rio Grande do Sul Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre Porto Alegre Rio Grande do Sul Brazil; Immunobiology and Immunogenetics Laboratory, Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil; Postgraduate Program in Medicine, Medical Sciences, Federal University of Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil.
| |
Collapse
|
30
|
Sasaki H, Miyata J, Kawana A, Fukunaga K. Antiviral roles of eosinophils in asthma and respiratory viral infection. FRONTIERS IN ALLERGY 2025; 6:1548338. [PMID: 40083723 PMCID: PMC11903450 DOI: 10.3389/falgy.2025.1548338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Eosinophils are immune cells that are crucial for the pathogenesis of allergic diseases, such as asthma. These cells play multifunctional roles in various situations, including infection. They are activated during viral infections and exert antiviral activity. Pattern recognition receptors, toll-like receptor 7 and retinoic acid inducible gene-I, are important for the recognition and capture of RNA viruses. In addition, intracellular granule proteins (eosinophil cationic protein and eosinophil-derived neurotoxin) and intracellular nitric oxide production inactivate and/or degrade RNA viruses. Interestingly, eosinophil-synthesizing specialized pro-resolving mediators possess antiviral properties that inhibit viral replication. Thus, eosinophils may play a protective role during respiratory virus infections. Notably, antiviral activities are impaired in patients with asthma, and eosinophil activities are perturbed in proportion with the severity of asthma. The exact roles of eosinophils in RNA virus (rhinovirus, respiratory syncytial virus, and influenza virus)-induced type 2 inflammation-based asthma exacerbation remain unclear. Our research demonstrates that interferons (IFN-α and IFN-γ) stimulate human eosinophils to upregulate antiviral molecules, including guanylate-binding proteins and tripartite motifs. Furthermore, IFN-γ specifically increases the expression of IL5RA, ICAM-1, and FCGR1A, potentially enhancing cellular responsiveness to IL-5, ICAM-1-mediated adhesion to rhinoviruses, and IgG-induced inflammatory responses, respectively. In this review, we have summarized the relationship between viral infections and asthma and the mechanisms underlying the development of antiviral functions of human and mouse eosinophils in vivo and in vitro.
Collapse
Affiliation(s)
- Hisashi Sasaki
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Jun Miyata
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihiko Kawana
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Koichi Fukunaga
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
31
|
Nagy MZ, Plaza-Rojas LB, Boucher JC, Kostenko E, Austin AL, Tarhini AA, Chen Z, Du D, Ojwang' AME, Davis J, Obermayer A, Rejniak KA, Shaw TI, Guevara-Patino JA. Effector T cells under hypoxia have an altered transcriptome similar to tumor-stressed T cells found in non-responsive melanoma patients. J Immunother Cancer 2025; 13:e010153. [PMID: 40010774 PMCID: PMC12086921 DOI: 10.1136/jitc-2024-010153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/26/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND In the tumor microenvironment (TME), hypoxia stands as a significant factor that modulates immune responses, especially those driven by T cells. As T cell-based therapies often fail to work in solid tumors, this study aims to investigate the effects of hypoxia on T cell topo-distribution in the TME, gene expression association with T cell states, and clinical responses in melanoma. METHODS To generate detailed information on tumor oxygenation and T cell accessibility, we used mathematical modeling of human melanoma tissue microarrays that incorporate oxygen supply from vessels, intratumoral diffusion, and cellular uptake. We created tumor maps and derived plots showing the fraction of CD4 and CD8 T cells against the distance to the nearest vessel and oxygen pressure. To assess their function and transcriptional changes caused by hypoxia, effector T cells were generated and cultured under hypoxia (0.5% oxygen) or normoxia (21% oxygen). The T cell hypoxia-transcriptional signature was compared against datasets from msigDB, iATLAS (clinical trials of melanoma patients treated with immune checkpoint inhibitors (ICIs)), ORIEN AVATAR (real-world melanoma patients treated with ICIs), and a single-cell atlas of tumor-infiltrating lymphocytes. RESULTS We made three specific observations: (1) in melanoma T cells preferentially accumulated in oxygenated areas close to blood vessels (50-100 µm from the vasculature in the regions of high oxygen availability) but not in hypoxic areas far from blood vessels. (2) Our analysis confirmed that under hypoxia, T cell functions were significantly reduced compared with normoxic conditions and accompanied by a unique gene signature. Furthermore, this hypoxic gene signature was prevalent in resting and non-activated T cells. Notably and clinically relevant, the hypoxic T cell gene set was found to correlate with reduced overall survival and reduced progression-free survival in melanoma patients, which was more pronounced in non-responder patients undergoing ICI therapy. (3) Finally, compared with a single-cell atlas of tumor-infiltrating T cells, our hypoxia signature aligned with a population of cells at a state termed stress response state (TSTR). CONCLUSIONS Our study highlights the critical role of hypoxia in shaping T cell distribution and its correlation with clinical outcomes in melanoma. We revealed a preferential accumulation of T cells in oxygenated areas. Moreover, hypoxic T cells develop a distinct hypoxic gene signature prevalent in resting, non-activated T cells and TSTR that was also associated with poorer outcomes, particularly pronounced among non-responders to ICIs.
Collapse
Affiliation(s)
- Mate Z Nagy
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Lourdes B Plaza-Rojas
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Justin C Boucher
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Elena Kostenko
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Anna L Austin
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Ahmad A Tarhini
- Departments of Cutaneous Oncology and Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Zhihua Chen
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Dongliang Du
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Awino Maureiq E Ojwang'
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Joshua Davis
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Alyssa Obermayer
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Katarzyna A Rejniak
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Timothy I Shaw
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Jose A Guevara-Patino
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
32
|
Zhao CY, Song C, He HW, Huang XZ, Meng XY, Huang AC, Xu CY, Luo LL, Xi SY, Lan YQ, Li WW, Lin YR, Zhu QD. Clinical Characteristics Analysis of 30 Cases of Interferon-γ Autoantibody-Positive Patients with Concurrent Mycobacterial Infection: A 6-Year Retrospective Study. Infect Drug Resist 2025; 18:1097-1110. [PMID: 40027921 PMCID: PMC11871848 DOI: 10.2147/idr.s493956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/08/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose This study aimed to investigate and elucidate the clinical characteristics, immune status, infection types and patterns, treatment responses, and disease progression in patients with positive anti-interferon-gamma (IFN-γ) autoantibodies in combination with Mycobacterium infections. Patients and Methods We conducted a retrospective analysis of clinical data from patients with positive anti-IFN-γ autoantibodies and concurrent Mycobacterial infections, including Mycobacterial infections (MTB) and non-tuberculous mycobacteria (NTM). The study included cases treated at the Fourth People's Hospital of Nanning, Guangxi, from 2018 to 2023. Data collected comprised symptoms, clinical signs, laboratory test results, imaging findings, and other relevant clinical information. Patients were also followed up to evaluate treatment responses and long-term therapeutic outcomes. Results A total of 30 patients with MTB and NTM infections were analyzed. The majority presented with common symptoms, such as cough, sputum production, weight loss, extrapulmonary tuberculosis (TB), and a range of opportunistic infections. Laboratory and imaging studies revealed complex infection patterns and various pathological changes. Treatment primarily involved targeted anti-infective therapy combined with immunosupportive measures. However, frequent treatment relapses and side effects were observed, resulting in two deaths. Conclusion Immune deficiency associated with positive anti-IFN-γ autoantibodies resembles the immunosuppression seen in advanced stages of human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS), rendering patients highly susceptible to opportunistic infections. These infections were predominantly caused by NTM, followed by MTB and Talaromyces marneffei (TM). This represents a novel immune deficiency syndrome that predisposes patients to a spectrum of opportunistic infections.
Collapse
Affiliation(s)
- Chun-Yan Zhao
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
- Clinical Medical School, Guangxi Medical University, Nanning, People’s Republic of China
| | - Chang Song
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
- Clinical Medical School, Guangxi Medical University, Nanning, People’s Republic of China
| | - Hua-Wei He
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Xian-Zhen Huang
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Xia-Yan Meng
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Ai-Chun Huang
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Chao-Yan Xu
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Li-Li Luo
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Shao-Yong Xi
- Department of Clinical Laboratory, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Yan-Qun Lan
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Wei-Wen Li
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Yan-Rong Lin
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| | - Qing-Dong Zhu
- Department of Tuberculosis, The Fourth People’s Hospital of Nanning, Nanning, People’s Republic of China
| |
Collapse
|
33
|
Ponce-Regalado MD, Becerril-Villanueva E, Maldonado-García JL, Moreno-Lafont MC, Martínez-Ramírez G, Jacinto-Gutiérrez S, Arreola R, Sánchez-Huerta K, Contis-Montes de Oca A, López-Martínez KM, Bautista-Rodríguez E, Chin-Chan JM, Pavón L, Pérez-Sánchez G. Comprehensive view of suicide: A neuro-immune-endocrine approach. World J Psychiatry 2025; 15:98484. [PMID: 39974471 PMCID: PMC11758041 DOI: 10.5498/wjp.v15.i2.98484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/26/2024] [Accepted: 12/23/2024] [Indexed: 01/14/2025] Open
Abstract
Suicide is defined as the act of a person attempting to take their own life by causing death. Suicide is a complex phenomenon that is influenced by a multitude of factors, including psychosocial, cultural, and religious aspects, as well as genetic, biochemical, and environmental factors. From a biochemical perspective, it is crucial to consider the communication between the endocrine, immune, and nervous systems when studying the etiology of suicide. Several pathologies involve the bidirectional communication between the peripheral activity and the central nervous system by the action of molecules such as cytokines, hormones, and neurotransmitters. These humoral signals, when present in optimal quantities, are responsible for maintaining physiological homeostasis, including mood states. Stress elevates the cortisol and proinflammatory cytokines levels and alter neurotransmitters balance, thereby increasing the risk of developing a psychiatric disorder and subsequently the risk of suicidal behavior. This review provides an integrative perspective about the neurochemical, immunological, and endocrinological disturbances associated with suicidal behavior, with a particular focus on those alterations that may serve as potential risk markers and/or indicators of the state preceding such a tragic act.
Collapse
Affiliation(s)
- María D Ponce-Regalado
- Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos 47620, Jalisco, Mexico
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - José Luis Maldonado-García
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11350, Mexico
| | - Martha C Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11350, Mexico
| | - Gabriela Martínez-Ramírez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
- Facultad de Medicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional autónoma de México, Tlalnepantla 54090, Mexico
| | - Salomón Jacinto-Gutiérrez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Rodrigo Arreola
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Karla Sánchez-Huerta
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico
| | - Arturo Contis-Montes de Oca
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | | | | | - José Miguel Chin-Chan
- Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Campeche, Campeche 24039, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| |
Collapse
|
34
|
Gong S, Li Y, Yan K, Shi Z, Leng J, Bao Y, Ning K. The Crosstalk Between Endothelial Cells, Smooth Muscle Cells, and Macrophages in Atherosclerosis. Int J Mol Sci 2025; 26:1457. [PMID: 40003923 PMCID: PMC11855868 DOI: 10.3390/ijms26041457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease closely tied to cellular metabolism. Recent genome-wide association study data have suggested the significant roles of endothelial cells, smooth muscle cells, and macrophages in the regression and exacerbation of AS. However, the impact of cellular crosstalk and cellular metabolic derangements on disease progression in AS is vaguely understood. In this review, we analyze the roles of the three cell types in AS. We also summarize the crosstalk between the two of them, and the associated molecules and consequences involved. In addition, we emphasize potential therapeutic targets and highlight the importance of the three-cell co-culture model and extracellular vesicles in AS-related research, providing ideas for future studies.
Collapse
Affiliation(s)
- Sihe Gong
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yanni Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Kaijie Yan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Zhonghong Shi
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Jing Leng
- Preclinical Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China;
| | - Yimin Bao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| |
Collapse
|
35
|
Garcia-Fabiani MB, Haase S, Banerjee K, Zhu Z, McClellan BL, Mujeeb AA, Li Y, Tronrud CE, Varela ML, West ME, Yu J, Kadiyala P, Taher AW, Núñez FJ, Alghamri MS, Comba A, Mendez FM, Nicola Candia AJ, Salazar B, Nunez FM, Edwards MB, Qin T, Cartaxo RT, Niculcea M, Koschmann C, Venneti S, Vallcorba MP, Nasajpour E, Pericoli G, Vinci M, Kleinman CL, Jabado N, Chandler JP, Sonabend AM, DeCuypere M, Hambardzumyan D, Prolo LM, Mahaney KB, Grant GA, Petritsch CK, Welch JD, Sartor MA, Lowenstein PR, Castro MG. H3.3-G34R Mutation-Mediated Epigenetic Reprogramming Leads to Enhanced Efficacy of Immune Stimulatory Gene Therapy in Diffuse Hemispheric Gliomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.06.13.544658. [PMID: 37398299 PMCID: PMC10312611 DOI: 10.1101/2023.06.13.544658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Diffuse hemispheric glioma (DHG), H3 G34-mutant, representing 9-15% of cases, are aggressive Central Nervous System (CNS) tumors with poor prognosis. This study examines the role of epigenetic reprogramming of the immune microenvironment and the response to immune-mediated therapies in G34-mutant DHG. To this end, we utilized human G34-mutant DHG biopsies, primary G34-mutant DHG cultures, and genetically engineered G34-mutant mouse models (GEMMs). Our findings show that the G34 mutation alters histone marks' deposition at promoter and enhancer regions, leading to the activation of the JAK/STAT pathway, which in turn results in an immune-permissive tumor microenvironment. The implementation of Ad-TK/Ad-Flt3L immunostimulatory gene therapy significantly improved median survival, and lead to over 50% long term survivors. Upon tumor rechallenge in the contralateral hemisphere without any additional treatment, the long-term survivors exhibited robust anti-tumor immunity and immunological memory. These results indicate that immune-mediated therapies hold significant potential for clinical translation in treating patients harboring H3.3-G34 mutant DHGs, offering a promising strategy for improving outcomes in this challenging cancer subtype affecting adolescents and young adults (AYA). STATEMENT OF SIGNIFICANCE This study uncovers the role of the H3.3-G34 mutation in reprogramming the tumor immune microenvironment in diffuse hemispheric gliomas. Our findings support the implementation of precision medicine informed immunotherapies, aiming at improving enhanced therapeutic outcomes in adolescents and young adults harboring H3.3-G34 mutant DHGs.
Collapse
Affiliation(s)
- Maria B. Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Present address: Leloir Institute Foundation, Buenos Aires, Argentina
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ziwen Zhu
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brandon L. McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anzar A. Mujeeb
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yingxiang Li
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Claire E. Tronrud
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria L. Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Molly E.J. West
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jin Yu
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
- Present address: Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ayman W. Taher
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Felipe J. Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Flor M. Mendez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alejandro J. Nicola Candia
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brittany Salazar
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fernando M. Nunez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marta B. Edwards
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rodrigo T. Cartaxo
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Michael Niculcea
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Carl Koschmann
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Sriram Venneti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Emon Nasajpour
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Giulia Pericoli
- Department of Onco-Hematology, Gene and Cell Therapy, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Maria Vinci
- Department of Onco-Hematology, Gene and Cell Therapy, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Claudia L. Kleinman
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - James P. Chandler
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael DeCuypere
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Division of Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Laura M. Prolo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Gerald A. Grant
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
- Present address: Department of Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Claudia K Petritsch
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Joshua D. Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
36
|
Mebumroong S, Lin H, Jermsutjarit P, Tantituvanont A, Nilubol D. Field Investigation Evaluating the Efficacy of Porcine Reproductive and Respiratory Syndrome Virus Type 2 (PRRSV-2) Modified Live Vaccines in Nursery Pigs Exposed to Multiple Heterologous PRRSV Strains. Animals (Basel) 2025; 15:428. [PMID: 39943198 PMCID: PMC11815747 DOI: 10.3390/ani15030428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
This study was conducted to evaluate the protective efficacy of modified live vaccines (MLVs) against porcine reproductive and respiratory syndrome (PRRS) in nursery pigs in a worst case scenario where MLV does not match the genetic profile of the field isolate, different MLVs are used for sows and piglets, and piglets are naturally exposed to genetically distinct heterologous PRRS virus (PRRSV) isolates. We divided 76,075, 2-week-old piglets from a seropositive sow herd vaccinated with US1-MLV into four groups. US1-MLV, US2-MLV, and US3-MLV groups were vaccinated with PRRSV-2 MLV including Ingelvac® PRRS MLV (Boehringer Ingelheim, Ingelheim am Rhein, Germany), HP-PRRSV-2 based MLV (Harbin Veterinary Research Institute, CAAS, Harbin, China), and Prime Pac® PRRS (MSD Animal Health, Rahway, NJ, USA), respectively. The NonVac group was left unvaccinated. At 0, 14, 28, and 56 days post-vaccination (DPV), sera were assayed for the presence of PRRSV-specific antibodies using ELISA and serum neutralization (SN), and PRRSV RNA using PCR. Average daily gain (ADG) and survival rates were compared between treatment groups. The results demonstrated vaccinated groups significantly improved in ADG compared to the non-vaccinated control group. Only US1-MLV and US3-MLV were able to significantly reduce mortality associated with field PRRSV infection in nursery pigs. Pigs vaccinated with US3-MLV displayed significantly lower mortality and higher ADG compared to all other groups. Field isolates were isolated and genetically compared to all three MLV vaccines at the start of the trial. The MLV with closest genetic similarity to the field isolate was US2-MLV by ORF5 gene comparison. This provided the lowest protection judging by ADG improvement and mortality reduction, as compared to US1-MLV and US3-MLV. Separately, strains of Thai PRRSV-2 isolates collected in 2017, 2019, and 2020 in the study area were investigated for evolutionary changes. Over time, we observed a shift in PRRSV-2 isolates from lineage 8.7 to lineage 1. The field isolates found shared 82.59-84.42%, 83.75-85.74%, and 84.25-85.90% nucleotide identity with the US1-MLV, US3-MLV and US2-MLV based vaccine, respectively. Our findings suggest genetic similarity between field viruses and vaccine strains should not be used as a predictor of field performance. We found that zootechnical performance of piglets was best in US3-MLV, despite sows being treated with a different vaccine The results also support that different MLVs can be used at different stages of production. Finally, we concluded that the shift from lineage 8.7 to lineage 1 was due to shifts in the worldwide prevalence of PRRSV isolates during that period of time and not due to vaccine recombination between isolates. Overall, MLV vaccine selection should be based on production performance and safety profile.
Collapse
Affiliation(s)
- Sunit Mebumroong
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand; (S.M.); (P.J.)
| | - Hongyao Lin
- MSD Animal Health Innovation Pte Ltd., Perahu Road, Singapore 718847, Singapore;
| | - Patumporn Jermsutjarit
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand; (S.M.); (P.J.)
| | - Angkana Tantituvanont
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Dachrit Nilubol
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand; (S.M.); (P.J.)
| |
Collapse
|
37
|
Lauterbach‐Rivière L, Thuringer L, Feld P, Toews LK, Schüssler J, Klinz J, Gläser L, Lohse S, Sternjakob A, Gasparoni G, Kattler‐Lackes K, Walter J, Lauterbach MA, Rahmann S, Möller L, Laue M, Janssen M, Stöckle M, Schmit D, Fliser D, Smola S. Tumor Necrosis Factor-Alpha Inhibits the Replication of Patient-Derived Archetype BK Polyomavirus While Activating Rearranged Strains. J Med Virol 2025; 97:e70210. [PMID: 39949253 PMCID: PMC11826303 DOI: 10.1002/jmv.70210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025]
Abstract
To date, no drugs are approved for BK polyomavirus (BKPyV) reactivation, a major cause of nephropathy after kidney transplantation. Recently, tumor necrosis factor-α (TNF-α) blockade has been proposed as a promising therapy, however, the effect of TNF-α on the clinically most common archetype (ww) BKPyV remained unclear. Assays in primary renal proximal tubule epithelial cells (RPTEC) allowed efficient replication only of BKPyV strains with rearranged (rr) non-coding control regions (NCCR), which may develop at later disease stages, but not of ww-BKPyV. Here, we optimized culture conditions allowing robust replication of patient-derived ww-BKPyV, while efficiently preserving their ww-NCCR. TNF-α promoted rr-BKPyV replication, while the TH1 cytokine IFN-γ suppressed it, also in the presence of TNF-α. Surprisingly, TNF-α alone was sufficient to suppress all ww-BKPyV strains tested. Comprehensive analysis using siRNAs, and chimeric or mutated BKPyV-strains revealed that the response to TNF-α depends on the NCCR type, and that the NF-κB p65 pathway but not the conserved NF-κB binding site is essential for the TNF-α-induced enhancement of rr-BKPyV replication. Our data suggest that in immunosuppressed patients with archetype-dominated infections, TNF-α blockade could interfere with natural TNF-α-mediated anti-BKPyviral control, and this could be detrimental when IFN-γ-driven TH1 responses are impaired. Ongoing inflammation, however, could lead to the selection of rearrangements responding to NCCR-activating pathways downstream of NF-κB p65 signaling, that may overcome the initial TNF-α-mediated suppression. Our findings also highlight the importance of using clinically relevant BKPyV isolates for drug testing and discovery, for which this new assay paves the way.
Collapse
Affiliation(s)
| | - Lucia Thuringer
- Institute of VirologySaarland University Medical CenterHomburgGermany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection ResearchSaarland University CampusSaarbrückenGermany
| | - Pascal Feld
- Institute of VirologySaarland University Medical CenterHomburgGermany
| | | | - Jessica Schüssler
- Institute of VirologySaarland University Medical CenterHomburgGermany
| | - Jonas Klinz
- Institute of VirologySaarland University Medical CenterHomburgGermany
| | - Lars Gläser
- Institute of VirologySaarland University Medical CenterHomburgGermany
| | - Stefan Lohse
- Institute of VirologySaarland University Medical CenterHomburgGermany
| | - Anna Sternjakob
- Institute of VirologySaarland University Medical CenterHomburgGermany
| | | | | | - Jörn Walter
- Department of GeneticsSaarland UniversitySaarbrückenGermany
| | - Marcel A. Lauterbach
- Molecular Imaging, Center for Integrative Physiology and Molecular MedicineSaarland UniversityHomburgGermany
| | - Sven Rahmann
- Algorithmic Bioinformatics, Center for Bioinformatics Saar, Saarland Informatics CampusSaarland UniversitySaarbrückenGermany
| | - Lars Möller
- Advanced Light and Electron Microscopy, Centre for Biological Threats and Special Pathogens, Robert Koch InstituteBerlinGermany
| | - Michael Laue
- Advanced Light and Electron Microscopy, Centre for Biological Threats and Special Pathogens, Robert Koch InstituteBerlinGermany
| | - Martin Janssen
- Department of UrologySaarland University Medical CenterHomburgGermany
| | - Michael Stöckle
- Department of UrologySaarland University Medical CenterHomburgGermany
| | - David Schmit
- Department of NephrologySaarland University Medical CenterHomburgGermany
| | - Danilo Fliser
- Department of NephrologySaarland University Medical CenterHomburgGermany
| | - Sigrun Smola
- Institute of VirologySaarland University Medical CenterHomburgGermany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection ResearchSaarland University CampusSaarbrückenGermany
| |
Collapse
|
38
|
Natura G, Vazquez E, Richter F, Segond von Banchet G, Ebbinghaus M, Ebersberger A, König C, Maltritz J, Gajda M, Schmidt-Hieber C, Schaible HG. Antinociceptive interactions between excitatory interferon-γ and interleukin-17 in sensory neurons. Brain Behav Immun 2025; 124:55-73. [PMID: 39566665 DOI: 10.1016/j.bbi.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024] Open
Abstract
Interferon-γ (IFNγ) and interleukin-17 (IL-17) are master regulators of innate and adaptive immunity. Here we asked whether these cytokines also regulate pain. Both cytokines increased the excitability of isolated small- to medium-sized sensory neurons, suggesting a pronociceptive effect. However, in vivo IL-17 was pronociceptive, whereas IFNγ was antinociceptive. Co-administration of IFNγ and IL-17 in vivo resulted in antinociception. Pre-incubation with IFNγ also eliminated the increase in excitability by interleukin-17A in isolated sensory neurons, demonstrating that the excitatory membrane effects of IFNγ can interfere with the excitatory membrane effects of IL-17, resulting in neuronal inhibition. IFNγ increased TTX-sensitive Na+ currents, while IL-17 increased TTX-resistant Na+ currents. Blocking TTX-sensitive Na+ currents eliminated the inhibition of the IL-17 effect by IFNγ. We propose a novel form of inhibition in sensory neurons that allows the intrinsically excitatory IFNγ to attenuate pro-nociceptive effects of cytokines such as IL-17 through interactions with voltage-gated Na+ currents.
Collapse
Affiliation(s)
- Gabriel Natura
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Enrique Vazquez
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Frank Richter
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Gisela Segond von Banchet
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Matthias Ebbinghaus
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Andrea Ebersberger
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Christian König
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Jakob Maltritz
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Mieczyslaw Gajda
- Institute of Legal Medicine, Section Pathology, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
| | - Christoph Schmidt-Hieber
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Hans-Georg Schaible
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany.
| |
Collapse
|
39
|
Xu M, Yang Z, Yang N, Li H, Ma H, Yi J, Hou H, Han F, Ma Z, Chen C. Development and Immunogenicity Study of Subunit Vaccines Based on Spike Proteins of Porcine Epidemic Diarrhea Virus and Porcine Transmissible Gastroenteritis Virus. Vet Sci 2025; 12:106. [PMID: 40005866 PMCID: PMC11860644 DOI: 10.3390/vetsci12020106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/15/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) are responsible for significant economic losses in the swine industry. The S1 proteins of these viruses serve as key targets for vaccine development. In this study, prokaryotic expression vectors for pCZN1-PEDV S1, pCZN1-TGEV S1, and pCZN1-PEDV S1-TGEV S1 were constructed. The corresponding proteins were expressed, purified, and used to prepare monovalent, bivalent, and mixed (PEDV S1 + TGEV S1) vaccines. Kunming (KM) mice were immunized with subunit vaccines, with PBS as the negative control (NC) and a commercial inactivated vaccine as the positive control (PC). Immune responses, including specific antibody (IgG, IgG1, IgG2a) levels, virus neutralization, and IFN-γ production, were evaluated. All vaccines induced high levels of specific IgG, IgG1, and IgG2a antibodies. At weeks 2 and 8, the PEDV S1 + TGEV S1 vaccine induced significantly higher levels of specific IgG and IgG1 compared to the PC (p < 0.001). The PEDV S1 vaccine also induced significantly higher specific IgG2a levels than the PC at week 4 (p < 0.0001). Virus neutralization assays demonstrated that the subunit vaccines induced neutralizing antibody levels comparable to or exceeding those of the PC. Furthermore, IFN-γ levels were significantly elevated in all vaccinated groups compared to the NC (p < 0.0001), indicating a robust immune response. These results suggest that the subunit vaccines are promising candidates for the safe and effective control of both PEDV and TGEV infections.
Collapse
Affiliation(s)
- Mingguo Xu
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Zhonglian Yang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Ningning Yang
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang 464000, China;
| | - Honghuan Li
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Hailong Ma
- Department of Biotechnology, Linxia Modern Career Academy, Linxia 731100, China;
| | - Jihai Yi
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Huilin Hou
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Fangfang Han
- The College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China;
| | - Zhongchen Ma
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| |
Collapse
|
40
|
Alvarado-Harris R, Perreira K, Woods-Giscombe CL, Mills‐Koonce WR, Santos HP. Prenatal inflammation and trauma symptoms in Latina mothers: The role of discrimination and growing up in an ethnic minoritized context. Brain Behav Immun Health 2025; 43:100914. [PMID: 39677061 PMCID: PMC11638603 DOI: 10.1016/j.bbih.2024.100914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
Background The race-based traumatic stress model proposes that discrimination elicits trauma-related symptoms. Cumulative discriminatory experiences and subsequent trauma symptoms may lead to prenatal inflammation, with far reaching consequences for the health of a mother and her child. Methods Latina mothers, primarily of Mexican and Central American heritage (n = 150), completed the Everyday Discrimination Scale and the Traumatic Avoidance subscale of the Inventory of Depression and Anxiety Symptoms-II during pregnancy (24-32 weeks). Plasma levels of cytokines were measured with multiplex assays, which were aggregated into a pro-inflammatory cytokine profile (IL-1β, TNF-α, IFN-γ, and IL-8) after a Confirmatory Factor Analysis supported this approach. Results Latina mothers who grew up in the US reported more discrimination, more traumatic avoidance symptoms, and had a more elevated cytokine profile than those who immigrated after childhood. Based on a two-mediator sequential model, discrimination and traumatic avoidance symptoms sequentially provided mechanistic support for the higher levels of cytokines observed in mothers who grew up in the US. Additionally, mothers who experienced trauma symptoms in response to discrimination had an elevated cytokine profile, whereas those who did not had a suppressed cytokine profile. Conclusion This is among the first studies to examine the association between trauma symptoms, discrimination, and inflammation during pregnancy. In so doing, it elucidates critical pathways by which discrimination may be differentially biologically embedded across immigrant generations. Emotional responses to and chronicity of discrimination may be critical factors for understanding how experiences of discrimination may influence the maternal inflammatory milieu.
Collapse
Affiliation(s)
- Rebeca Alvarado-Harris
- School of Nursing, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Krista Perreira
- Department of Social Medicine, University of North Carolina at Chapel Hill, North Carolina, United States
| | | | | | - Hudson P. Santos
- The University of Miami School of Nursing and Health Studies, Florida, United States
| |
Collapse
|
41
|
Clark DN, Brown SV, Xu L, Lee RL, Ragusa JV, Xu Z, Milner JD, Filiano AJ. Prolonged STAT1 signaling in neurons causes hyperactive behavior. Brain Behav Immun 2025; 124:1-8. [PMID: 39542073 PMCID: PMC11745914 DOI: 10.1016/j.bbi.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/29/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
The interferon (IFN)-induced STAT1 signaling pathway is a canonical immune pathway that has also been implicated in regulating neuronal activity. The pathway is enriched in brains of individuals with autism spectrum disorder (ASD) and schizophrenia (SZ). Over-activation of the STAT1 pathway causes pathological transcriptional responses, however it is unclear how these responses might translate into behavioral phenotypes. We hypothesized that prolonged STAT1 signaling in neurons would be sufficient to cause behavioral deficits associated with neurodevelopmental disorders. In this study, we developed a novel mouse model with the clinical STAT1 gain-of-function mutation, T385M, in neurons. These mice were hyperactive and displayed neural hypoactivity with less neuron counts in the caudate putamen. Driving the STAT1 gain-of-function mutation exclusively in dopaminergic neurons, which project to the caudate putamen of the dorsal striatum, mimicked some hyperactive behaviors without a reduction of neurons. Moreover, we demonstrated that this phenotype is neuron specific, as mice with prolonged STAT1 signaling in all excitatory or inhibitory neurons or in microglia were not hyperactive. Overall, these findings suggest that STAT1 signaling in neurons is a crucial player in regulating striatal neuron activity and aspects of motor behavior.
Collapse
Affiliation(s)
- Danielle N Clark
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA; Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Shelby V Brown
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Li Xu
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Rae-Ling Lee
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Joey V Ragusa
- Department of Pathology, Duke University, Durham, NC, USA
| | - Zhenghao Xu
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Joshua D Milner
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Anthony J Filiano
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA; Marcus Center for Cellular Cures, Duke University, Durham, NC, USA; Department of Pathology, Duke University, Durham, NC, USA; Department of Neurosurgery, Duke University, Durham, NC, USA.
| |
Collapse
|
42
|
Doghish AS, Elazazy O, Mohamed HH, Mansour RM, Ghanem A, Faraag AHI, Elballal MS, Elrebehy MA, Elesawy AE, Abdel Mageed SS, Saber S, Nassar YA, Abulsoud AI, Abdel-Reheim MA, Elawady AS, Ali MA, Basiouny MS, Hemdan M, Lutfy RH, Awad FA, El-Sayed SA, Ashour MM, El-Sayyad GS, Mohammed OA. A Review on miRNAs in Enteric Bacteria-mediated Host Pathophysiology: Mechanisms and Implications. J Biochem Mol Toxicol 2025; 39:e70160. [PMID: 39907181 DOI: 10.1002/jbt.70160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/22/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025]
Abstract
Recently, many studies focused on the billions of native bacteria found inside and all over the human body, commonly known as the microbiota, and its interactions with the eukaryotic host. One of the niches for such microbiota is the gastrointestinal tract (GIT), which harbors hundreds to thousands of bacterial species commonly known as enteric bacteria. Changes in the enteric bacterial populations were linked to various pathologies such as irritable bowel syndrome and obesity. The gut microbiome could affect the health status of individuals. MicroRNAs (miRNAs) are one of the extensively studied small-sized noncoding RNAs (ncRNAs) over the past decade to explore their multiple roles in health and disease. It was proven that miRNAs circulate in almost all body fluids and tissues, showing signature patterns of dysregulation associated with pathologies. Both cellular and circulating miRNAs participate in the posttranscriptional regulation of genes and are considered the potential key regulators of genes and participate in cellular communication. This manuscript explores the unique interplay between miRNAs and enteric bacteria in the gastrointestinal tract, emphasizing their dual role in shaping host-microbiota dynamics. It delves into the molecular mechanisms by which miRNAs influence bacterial colonization and host immune responses, linking these findings to gut-related diseases. The review highlights innovative therapeutic and diagnostic opportunities, offering insights for targeted treatments of dysbiosis-associated pathologies.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Hend H Mohamed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
- Biochemistry Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Reda M Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Molecular Biology and Biotechnology Department, School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Ahmed H I Faraag
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Medical Department, School of Biotechnology, Badr University in Cairo, Badr City, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Galala University, New Galala City, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Yara A Nassar
- Department of Botany, Faculty of Science, Biotechnology and Its Application Program, Mansoura University, Mansoura, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
- Department of Biochemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Egypt
| | | | - Alaa S Elawady
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A Ali
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | | | - Mohamed Hemdan
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Radwa H Lutfy
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Farah A Awad
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Salma A El-Sayed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Mohamed M Ashour
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Gharieb S El-Sayyad
- Medical Laboratory Technology Department, Faculty of Applied Health Sciences Technology, Badr University in Cairo (BUC), Cairo, Egypt
- Microbiology and Immunology Department, Faculty of Pharmacy, Galala University, New Galala city, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
43
|
Chen JW, Wang YX, Gao RR, Ma LY, Zhong J, Yang JX, Deng ZH, Li YY, Li XL, Shu YH, Guo WJ, Zhou ZY, Tian XY, Ma J, Liu Y, Chen Q. CDK14 regulates the development and repair of lung. Cell Death Discov 2025; 11:12. [PMID: 39827158 PMCID: PMC11743204 DOI: 10.1038/s41420-025-02292-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/12/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Cyclin-dependent kinases (CDK) 14 regulates cell cycle, tumor expansion by influencing the downstream targets of the canonical Wnt signaling pathway. However, the function of CDK14 during organ development and regeneration has not been investigated in genetically-modified animals. Here, we found that genetic ablation of Cdk14 influenced pulmonary vascular endothelial cells and alveolar epithelial cells during mice embryonic development as well as repair of lung after bleomycin or lipopolysaccharide induced injury. Genetic knockout of Cdk14 and the CDK14 covalent inhibitor FMF-04-159-2 resulted in reduction of pulmonary vessel covered area and epithelial cell number, exhibiting increased mortality and more severe lung damage after injury. Mechanistically, Cdk14 ablation inhibited the proliferation of epithelial and vascular endothelial cells, inducing cell cycle arrest at the G2/M phase. Through RNA-seq analysis of both endothelial and epithelial cells, we found that knockdown of Cdk14 controls the expression of signal transducers and activator of transcription 1 (STAT1) as well as associated genes in interferon signaling. Disruption of Cdk14 interferes with IFN-γ induced lung repair in vivo, suggesting potential crosstalk of CDK14 signaling and IFN-γ pathway. Our work highlights the importance of Cdk14 in lung development and regenerative repair through an uncharacterized CDK14- IFN-γ signaling axis.
Collapse
Grants
- 32270866, 32470868, 32300693, 32471155, 22107045 National Natural Science Foundation of China (National Science Foundation of China)
- National Key R&D Program of China (2022YFA1103200); the Fundamental Research Funds for the Central Universities (2024ZYGXZR077); Guangzhou basic and applied basic research funding (2024A04J6259); The Pearl River Talent Recruitment Program (2023ZT10Y154, 2021ZT09Y233, 2023QN10Y147); South China University of Technology (D6241240); Talent Program and Basic Research Project of Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (1103792101, GIBHBRP23-02, GIBHBRP24-01); Cooperation Fund of CHCAMS and SZCH (CFA202201006); National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital,Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen (E010221005); Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences. (KLRB202201, KLRB202305); and partially supported by Science and Technology Planning Project of Guangdong Province, China (2023B1212060050,2023B1212120009).
Collapse
Affiliation(s)
- Jian-Wei Chen
- Institutes of physical science and information technology, Anhui University, Hefei, Anhui, 230601, China
| | - Yu-Xiang Wang
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510530, China
| | - Rong-Rong Gao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences; NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China
| | - Lan-Yue Ma
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510530, China
| | - Jing Zhong
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510530, China
| | - Jia-Xin Yang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zhao-Hua Deng
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510530, China
| | - Yu-Yan Li
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510530, China
| | - Xiao-Ling Li
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510530, China
| | - Ya-Hai Shu
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
| | - Wen-Jing Guo
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
| | - Zi-Yuan Zhou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xiao Yu Tian
- CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, Heart and Vascular Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| | - Jinjin Ma
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- The Institute of Future Health, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| | - Qi Chen
- Institutes of physical science and information technology, Anhui University, Hefei, Anhui, 230601, China.
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China.
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510530, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences; NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China.
| |
Collapse
|
44
|
Champagne J, Nielsen MM, Feng X, Montenegro Navarro J, Pataskar A, Voogd R, Giebel L, Nagel R, Berenst N, Fumagalli A, Kochavi A, Lovecchio D, Valcanover L, Malka Y, Yang W, Laos M, Li Y, Proost N, van de Ven M, van Tellingen O, Bleijerveld OB, Haanen JBAG, Olweus J, Agami R. Adoptive T cell therapy targeting an inducible and broadly shared product of aberrant mRNA translation. Immunity 2025; 58:247-262.e9. [PMID: 39755122 DOI: 10.1016/j.immuni.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/14/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025]
Abstract
Prolonged exposure to interferon-gamma (IFNγ) and the associated increased expression of the enzyme indoleamine 2,3-dioxygenase 1 (IDO1) create an intracellular shortage of tryptophan in the cancer cells, which stimulates ribosomal frameshifting and tryptophan to phenylalanine (W>F) codon reassignments during protein synthesis. Here, we investigated whether such neoepitopes can be useful targets of adoptive T cell therapy. Immunopeptidomic analyses uncovered hundreds of W>F neoepitopes mainly presented by the HLA-A∗24:02 allele. We identified a T cell receptor (TCRTMBIM6W>F.1) possessing high affinity and specificity toward TMBIM6W>F/HLA-A∗24:02, the inducible W>F neoepitope with the broadest expression across cancer cell lines. TCRTMBIM6W>F.1 T cells are activated by tryptophan-depleted cancer cells but not by non-cancer cells. Finally, we provide in vivo proof of concept for clinical application, whereby TCRMART1 T cells promote cancer cell killing by TCRTMBIM6W>F.1 T cells through the generation of W>F neoepitopes. Thus, neoepitopes arising from W>F substitution present shared and highly expressed immunogenic targets with the potential to overcome current limitations in adoptive T cell therapy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Animals
- Mice
- Protein Biosynthesis
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Cell Line, Tumor
- Tryptophan/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- RNA, Messenger/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/genetics
- Interferon-gamma/metabolism
- Antigens, Neoplasm/immunology
Collapse
Affiliation(s)
- Julien Champagne
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Morten M Nielsen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Xiaodong Feng
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jasmine Montenegro Navarro
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Abhijeet Pataskar
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rhianne Voogd
- Department of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lisanne Giebel
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Remco Nagel
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nadine Berenst
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Amos Fumagalli
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Adva Kochavi
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Domenica Lovecchio
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lorenzo Valcanover
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Yuval Malka
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Weiwen Yang
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Maarja Laos
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Yingqian Li
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Natalie Proost
- Preclinical Intervention Unit and Pharmacology Unit of the Mouse Clinic for Cancer and Ageing (MCCA), the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marieke van de Ven
- Preclinical Intervention Unit and Pharmacology Unit of the Mouse Clinic for Cancer and Ageing (MCCA), the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Onno B Bleijerveld
- NKI Proteomics facility, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B A G Haanen
- Department of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Reuven Agami
- Division of Oncogenomics, Oncode institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Erasmus MC, Department of Genetics, Rotterdam University, Rotterdam, the Netherlands.
| |
Collapse
|
45
|
Jeon SY, Shin HS, Lee H, Lee JO, Kim YS. The anti-tumor effect of the IFNγ/Fas chimera expressed on CT26 tumor cells. Anim Cells Syst (Seoul) 2025; 29:46-56. [PMID: 39777022 PMCID: PMC11703469 DOI: 10.1080/19768354.2024.2442393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/15/2024] [Accepted: 12/01/2024] [Indexed: 01/11/2025] Open
Abstract
Interferon gamma (IFNγ) is well-known for its ability to stimulate immune cells in response to pathogen infections and cancer. To develop an effective cancer therapeutic vaccine, CT26 colon carcinoma cells were genetically modified to express IFNγ either as a secreted form (sIFNγ) or as a membrane-bound form. For the membrane-bound expression, IFNγ was fused with Fas (mbIFNγ/Fas), incorporating the extracellular cysteine-rich domains, transmembrane, and cytoplasmic domains of Fas. The tumor cells expressing sIFNγ and mbIFNγ/Fas showed slower growth rates compared to the mock-transfected cells. Furthermore, the tumorigenicity of the CT26 cells expressing mbIFNγ/Fas was significantly lower than that of cells expressing sIFNγ or the mock control. Remarkably, about 85% of the mice injected with the mbIFNγ/Fas-expressing tumors remained tumor-free for over two months. Mice that rejected mbIFNγ/Fas-expressing tumors developed systemic anti-tumor immunity against CT26 cells, which was characterized by enhanced levels of CD4+ and CD8+ T cells, as well as natural killer (NK) cells. Interestingly, splenocytes activated with the mbIFNγ/Fas-expressing tumors exhibited higher cytotoxicity than those activated with tumor cells expressing sIFNγ. These findings suggest that expressing the mbIFNγ/Fas chimera in tumor cells could be a promising strategy for developing whole tumor cell vaccines or gene therapies for cancer immunotherapy.
Collapse
Affiliation(s)
- Seo Yeon Jeon
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Korea
| | - Hee-Su Shin
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Korea
| | - Hayyoung Lee
- Department of Life Sciences and Postech Biotech Center, POSTECH, Pohang, Korea
| | - Jie-Oh Lee
- Department of Life Sciences and Institute of Membrane Proteins, POSTECH, Pohang, Korea
| | - Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Korea
| |
Collapse
|
46
|
Ireland PA, Verheyden M, Jansson N, Sebaratnam D, Sullivan J. Infection risk with JAK inhibitors in dermatoses: a meta-analysis. Int J Dermatol 2025; 64:24-36. [PMID: 39367521 DOI: 10.1111/ijd.17501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 10/06/2024]
Abstract
Evolving evidence suggests that Janus Kinase Inhibitors (JAKi) may predispose to certain infections, including tuberculosis and human herpes viruses. This review aimed to compare the infection risk in patients on a systemic JAKi for a dermatologic indication to a placebo. A systematic review was carried out from inception to June 2023, using the EMBASE, Medline, SCOPUS, and Cochrane Library of Registered Trials databases. Eligible studies included placebo-controlled randomized trials that investigated the incidence of infection in patients with a dermatologic indication. Primary outcomes included the most commonly reported infections pertaining to serious and opportunistic infections, upper respiratory tract infections, nasopharyngitis, herpes simplex, varicella zoster, tuberculosis, neutropenia, and lymphopenia. A meta-analysis of incidence ratios was conducted to determine odds ratios (OR), with a 95% confidence interval (CI) analysis. The meta-analysis found no increased risk of serious (OR: 0.92, 95% CI: 0.61-1.43, P = 0.74) or opportunistic infections (OR: 0.65, 95% CI: 0.32-1.31, P = 0.23). The incidence of varicella-zoster infections was significantly higher in the JAKi cohort (OR: 1.72, 95% CI: 1.08-2.72, P = 0.022). From 25 studies, there was no overall increased risk of herpes simplex infections (OR: 1.43, 95% CI: 0.93-2.23, P = 0.102) to placebo; however, a significantly higher risk in those with atopic dermatitis to alopecia areata was demonstrated (OR: 1.73, 95% CI: 1.13-2.69, P = 0.013). The results of this analysis do not suggest an increased risk of serious and opportunistic infections in those on JAKi compared to placebo. However, they support an increased risk of varicella-zoster infections and a higher risk of herpes simplex infections in those with atopic dermatitis to alopecia areata. The results of this report support these agents' short-term safety but signal that vigilance should be practiced in patients at risk for serious or recurrent herpes virus infections.
Collapse
Affiliation(s)
- Patrick A Ireland
- Prince of Wales Hospital, Randwick, NSW, Australia
- University of new South Wales, Randwick, NSW, Australia
| | - Matthew Verheyden
- University of Sydney, Camperdown, NSW, Australia
- John Hunter Hospital, Newcastle, NSW, Australia
| | | | - Deshan Sebaratnam
- University of new South Wales, Randwick, NSW, Australia
- Liverpool Hospital, Liverpool, NSW, Australia
| | - John Sullivan
- University of new South Wales, Randwick, NSW, Australia
- Kingsway Dermatology and Aesthetics, Miranda, NSW, Australia
| |
Collapse
|
47
|
Abbasifard M, Ostad Ebrahimi H, Taghipur Khajeh Sharifi G, Bahrehmand F, Bagheri-Hosseinabadi Z. Investigation of the circulatory microRNAs and their involvement in regulation of inflammation in patients with COVID-19. Hum Immunol 2025; 86:111208. [PMID: 39667207 DOI: 10.1016/j.humimm.2024.111208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Dysregulated levels of cytokines may lead to cytokine storm, which has been implicated in the immunopathogenesis of coronavirus disease 2019 (COVID-19). Here in the current study, the role of microRNA (miR)-155-5p, miR-146a, and miR-221-3p in the regulation of the immune responses and inflammatory state in patients with COVID-19 was investigated. METHODS In this case-control study, peripheral blood samples were obtained from 75 COVID-19 subjects and 100 healthy controls. From the plasma samples, RNA was extracted and cDNA was synthesized, and subsequently the transcript level of miRNAs was measured by Real-time PCR. The plasma levels of interleukin (IL)-4 and interferon (IFN)-γ were determined using ELISA. RESULTS miR-155-5p (fold change = 1.87, P = 0.020) and miR-221-3p (fold change = 2.26, P = 0.008), but not miR-146a, was upregulated in the plasma sample of COVID-19 cases compared to controls. The level of IFN-γ (but not IL-4) was significantly higher in the plasma samples of COVID-19 patients compared to control group. The expression level of miR-155-5p (r = 0.35, corrected P = 0.066) and miR-221-3p (r = 0.25, corrected P = 0.066) had positive correlation with the plasma levels of IFN-γ. CONCLUSIONS IFN-γ pathway in involved in the pathogenesis of COVID-19 that is regulated through miR-155-5p and miR-221-3p. These miRNAs showed potential utility as biomarkers for predicting the severity of COVID-19.
Collapse
Affiliation(s)
- Mitra Abbasifard
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Ostad Ebrahimi
- Department of Paediatrics, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Fatemeh Bahrehmand
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Bagheri-Hosseinabadi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
48
|
Savitz J, McKinney BA, Meier TB, Zheng H, Ford BN, Yolken RH, Teague TK, Cole SW. Nuclear factor kappa-B cell (NF-κB), interferon regulatory Factor, and glucocorticoid receptor pathway activation in major depressive Disorder: The role of cytomegalovirus infection. Brain Behav Immun 2025; 123:1052-1060. [PMID: 39532200 PMCID: PMC11624063 DOI: 10.1016/j.bbi.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
Altered activity of major immunoregulatory pathways has been reported in major depressive disorder (MDD) and is thought to underlie the elevations in circulating inflammatory mediators present in a subgroup of patients. However, the drivers of these changes in gene expression remain unclear. One potential modulator of immune function is viral infection. Here we examined the relationship between cytomegalovirus (CMV), a common herpesvirus, that has been shown to be a pathological cofactor in inflammatory disorders, and activity of key coordinators of the innate inflammatory response in MDD. We used RNAseq to characterize gene expression differences in in 79 unmedicated individuals with MDD and 80 healthy controls (HCs). A well-established bioinformatic strategy was used to quantify transcription control pathway activity based on the relative prevalence of pre-specified transcription factor-binding motifs in the promoters of differentially expressed genes. The main aim was to characterize diagnostic differences in immunoregulatory pathway activity and determine if these were related to CMV serostatus or antibody titer (viral reactivation). Significantly increased activity of interferon regulatory factor 1 (IRF1) and nuclear factor kappa-B cell (NF-κB) pathways was observed in the MDD group compared with HCs. Transcript Origin Analyses using cell-specific reference transcriptomes indicated that the MDD-associated transcriptome changes derived primarily from myeloid lineage immune cells (classical and non-classical monocytes). A more modest MDD-associated upregulation of glucocorticoid receptor (GR) pathway activity was also present. CMV infection/activity across the combined MDD and HC groups was weakly related to GR pathway activation but not to IRF1 and NF-κB activity; the most salient signature of CMV was activation and/or expansion of the CD8+ T-cell population. The elevated MDD-associated NF-κB (but not IRF1) activity was markedly attenuated after controlling for CMV antibody titer or for CD8+ T-cell prevalence. At least some of the NF-κB signal in MDD may be attributable to the cellular immune response to CMV, suggesting that CMV infection may be one of several pathways contributing to inflammation in depression. The pronounced activation of the antiviral IRF-1 pathway in MDD suggests the contribution of viral processes although this specific antiviral effect was not specific to CMV.CMV may indirectly drive interferon responses by impairing T-cell control of other viral infections.
Collapse
Affiliation(s)
- Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa OK, USA; Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa OK, USA.
| | - Brett A McKinney
- Department of Mathematics and Computer Science, The University of Tulsa, Tulsa, OK, USA
| | - Timothy B Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI USA
| | - Haixia Zheng
- Laureate Institute for Brain Research, Tulsa OK, USA; Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa OK, USA
| | - Bart N Ford
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - T Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK, USA; Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK, USA; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Steve W Cole
- University of California, Los Angeles, Cousins Center for Psychoneuroimmunology, Los Angeles, CA, USA; University of California, Los Angeles, David Geffen School of Medicine, Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA, USA
| |
Collapse
|
49
|
Bamaga A, Kurdi M, Alkhotani A, Alghefari H, Gasemaltayeb R, Mrair F, Fadel ZT, Mehboob R, Tayyib A, Abuzinadah AR. Inflammatory myopathy with abundant macrophage [IMAM]: Systemic analysis and pathological approach to distinguish it from dermatomyositis. J Neuromuscul Dis 2025; 12:22143602251315376. [PMID: 39973451 DOI: 10.1177/22143602251315376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BACKGROUND Inflammatory myopathy with abundant macrophage [IMAM] is marked by macrophage infiltration and muscle fibers damage, resembling dermatomyositis [DM] but with unique pathology. Its mechanism remains unclear. Our study focused on exploring the clinicopathological characteristics, underlying pathogenic mechanisms, and the challenges in diagnosing and managing IMAM. METHODS A systematic analysis of medical literature databases (Pub-med, Cochrane, Scopus, Google Scholar) was performed using the term "IMAM," excluding studies on other inflammatory myopathies [IMs]. Selected studies were independently assessed with the Newcastle-Ottawa Scale, and quantitative data underwent inter-statistical analysis, descriptive and odds ratio, to identify relevant findings. RESULTS Eight studies, including 49 IMAM cases from 2003 to 2024, were analyzed. Five were case reports, and three were cross-sectional studies. IMAM showed no age or sex predilection. Common symptoms included proximal muscle weakness, pain, and fatigue, with atypical DM-like skin features in 65% of cases. Other association included hemophagocytosis, cutaneous panniculitis, and interstitial lung infiltration. Histologically, all cases showed myonecrosis infiltrated with CD68+ macrophages. Scattered CD3+ and CD4+ T-cells expressing IL-10 with no or rare CD8+ T-cells were identified. MAC deposition was limited to necrotic fibers, and perifascicular atrophy was absent in all cases. Anti-PL-7 and anti-U1 RNP antibodies were detected in 4% of cases. Elevated TNFα and IFN-γ levels, with low STAT1 and STAT6, were observed. Genetic analysis revealed MEFV polymorphisms in 7 cases and a TNFRSF1A mutation [C43R] in single case. Treatment involved steroids, with or without immunotherapy or chemotherapy, leading to remission and recovery in 43.7% of cases. CONCLUSION IMAM is a distinct type of IMs that requires muscle biopsy for diagnosis as myositis antibody and cytokine tests are usually insensitive.
Collapse
Affiliation(s)
- Ahmed Bamaga
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maher Kurdi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Alaa Alkhotani
- Department of Pathology, College of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Huda Alghefari
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Raed Gasemaltayeb
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Faisal Mrair
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Zahir T Fadel
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Awab Tayyib
- Department of Pathology, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Ahmad R Abuzinadah
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
50
|
Lu C, Liao S, Chen B, Xu L, Wu N, Lu D, Kang H, Zhang XB, Song G. Responsive probes for in vivo magnetic resonance imaging of nitric oxide. NATURE MATERIALS 2025; 24:133-142. [PMID: 39587281 DOI: 10.1038/s41563-024-02054-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024]
Abstract
Nitric oxide (NO), a pivotal signalling molecule, plays multifaceted roles in physiological and pathological processes, including cardiovascular and immune functions, neurotransmission and cancer progression. Nevertheless, measuring NO in vivo is challenging due to its transient nature and the complexity of the biological environment. Here we describe NO-responsive magnetic probes made of crosslinked superparamagnetic iron oxide nanoparticles tethered to a NO-sensitive cleavable linker for highly sensitive and selective NO magnetic resonance imaging in vivo. These probes enable the detection of NO at concentrations as low as 0.147 μM, allowing for the imaging and quantification of NO in mouse tumour models, studying its effects on tumour progression and immunity and assessing the response of tumour-associated macrophages to cancer immunotherapeutic agents. Additionally, they facilitate concurrent anatomical and molecular imaging of organs, helping to identify pathological alterations in the liver. Overall, these probes represent promising non-invasive tools for investigating the dose-dependent conflicting role of NO in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Chang Lu
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Shiyi Liao
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Baode Chen
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Li Xu
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Na Wu
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Dingyou Lu
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, Korea
| | - Xiao-Bing Zhang
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China.
| | - Guosheng Song
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China.
| |
Collapse
|