1
|
Kumar RR, Agarwal N, Shree A, Gorain JK, Rahul E, Ganguly S, Bakhshi S, Sharma U. Decoding the immune landscape in Ewing sarcoma pathogenesis: The role of tumor infiltrating immune cells and immune milieu. J Bone Oncol 2025; 52:100678. [PMID: 40242222 PMCID: PMC12002756 DOI: 10.1016/j.jbo.2025.100678] [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: 02/03/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Ewing sarcoma (EwS) is the second most prevalent pediatric bone malignancy, characterized by its aggressive behavior and unfavorable prognosis. The tumor microenvironment (TME) of EwS is shaped by immunosuppressive components, including myeloid-derived suppressor cells, tumor-associated macrophages, and immune checkpoint molecules such as PD-1/PD-L1 and HLA-G. These elements impair anti-tumor immune responses by modulating the function of tumor-infiltrating immune cells, such as regulatory T cells (Tregs), CD8+ T cells, and natural killer cells. Chemokines, including CXCL9 and CXCL12, and cytokines, such as transforming growth factor-beta and interleukin-10, further contribute to immune suppression and promote metastatic dissemination. Recent advances in immunotherapy have highlighted the therapeutic potential of modulating immune cells and signaling pathways to enhance anti-tumor immunity. This review provides a comprehensive analysis of the complex immune landscape within the EwS TME, focusing on the mechanistic roles of key immune components and their potential as therapeutic targets. Understanding these interactions could pave the way for innovative treatment strategies to improve clinical outcomes in patients with EwS.
Collapse
Affiliation(s)
- Rajiv Ranjan Kumar
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Nikita Agarwal
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Akshi Shree
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
- Department of Biomedical Science, Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, Delhi 110096, India
| | - Jaya Kanta Gorain
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Ekta Rahul
- Department of Pathology, Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, Delhi 110001, India
| | - Shuvadeep Ganguly
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Uttam Sharma
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
2
|
Wang YX, Ma YD, Li HH, Duo WJ, Jin QW, Zhou KJ, Gao YR, He JN, Xie YJ, Chu L, Yang XD. Schistosoma japonicum cystatin attenuated CLP-induced sepsis in mice though inducing tolerogenic dendritic cells and regulatory T cells. Comp Immunol Microbiol Infect Dis 2025; 120:102345. [PMID: 40344985 DOI: 10.1016/j.cimid.2025.102345] [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: 03/03/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Sepsis is a life-threatening complication caused by the overwhelming immune response to bacterial infection leading to the fatal organ damage and even death. Helminth infections modulate host's immune system through secreting functional proteins to reduce host immune attack as a survival strategy, therefore have been used for the therapy of some inflammatory or autoimmune diseases. Sj-Cys is a cysteine protease inhibitor secreted by Schistosoma japonicum exerting strong immunomodulatory function which has been used to treat sepsis, however, the mechanism underlying the therapeutic efficacy has not been fully elucidated. In this study, we expressed Sj-Cys as recombinant protein (rSj-Cys) in prokaryotic system and rSj-Cys was used to incubate with mouse bone marrow derived dendritic cells (BMDCs) in vitro. Our study revealed that rSj-Cys was able to induce differentiation of BMDCs to tolerant property (TolDCs). Adoptive transfer of rSj-Cys induced-TolDCs into mice with cecal ligation and puncture (CLP)-induced sepsis conferred a significant therapeutic effect on CLP-induced sepsis in mice with reduced mortality and vital organ damage. The therapeutic effect of Sj-Cys-induced TolDCs was associated with upregulation of CD3+CD4+CD25+Foxp3+ regulatory T cells (Tregs) and reduced inflammatory cytokines IL-6 and TNF-α and boosted level of regulatory cytokines IL-10 and TGF-β. The results identified in this study further suggest rSj-Cys has the potential to be developed into a drug substance for the treatment of inflammatory or autoimmune diseases due to its immunomodulatory effect on tolerant dendritic cells and regulatory T cells.
Collapse
Affiliation(s)
- Yi-Xiang Wang
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| | - Yi-Dan Ma
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| | - Hui-Hui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| | - Wen-Juan Duo
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China; Nan Jing Zi Jin Hospital, Nan Jin 210007, China.
| | - Qi-Wang Jin
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| | - Kai-Jun Zhou
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China.
| | - Yan-Ran Gao
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China.
| | - Jun-Nan He
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China.
| | - Yu-Jie Xie
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China.
| | - Liang Chu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China; Second Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China.
| | - Xiao-Di Yang
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| |
Collapse
|
3
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [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: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
4
|
Le ACT, Fiuza-Fernandes J, Silva JM, Sampaio MT, Texeira-Castro A, Duarte-Silva S, Leite-Almeida H. Pretreatment with dimethyl fumarate prevents chronic pain and its comorbidities via Nrf2 pathway in a rat model of neuropathic pain. Brain Behav Immun 2025; 128:725-736. [PMID: 40349732 DOI: 10.1016/j.bbi.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/24/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Chronic pain susceptibility varies across individuals, and the immune system, among other factors, appears to be involved in this variability. This study aims to examine whether modulating the immune system around the inciting events provides protection against the development of chronic pain and related comorbidities. Dimethyl fumarate (DMF) - an immunomodulator - or vehicle (VEH) was administered 7 days before spared nerve injury (SNI) in Wistar Han male rats. After the surgery, half of the animals from each group shifted treatments for an additional 7 days, resulting in 4 groups: continuous treatment (DMF-DMF), pretreatment (DMF-VEH), early treatment (VEH-DMF), and control (VEH-VEH). As a result, DMF-DMF, DMF-VEH, and VEH-DMF reduced allodynia and attenuated anhedonia measured at 4 weeks post-SNI, compared to VEH-VEH. VEH-DMF increased serum protein levels of anti-inflammatory markers, including IL-10 and G-CSF, and those pleiotropic, such as IL-6, IFN-g, and IL-17A. The leptin reduction seems to be a delayed effect of DMF. Meanwhile, other anti-inflammatory cytokines, IL-4 and IL-13, increased with relatively large effect sizes. The subsequent experiment shows that DMF pretreatment also alleviated anxiety- and cognitive dysfunction-like behaviors, measured at 6-7 weeks post-surgery. Additionally, this treatment significantly diminished SNI-induced protein ATF-3 - a neuronal injury marker - in L4-6 DRG at day 49 post-surgery. Nuclear factor E2-related factor 2 (Nrf2), a major regulator of the antioxidant and anti-inflammatory response, appears to be linked with DMF protective mechanisms, with trigonelline, an Nrf2 inhibitor, largely abolishing its effects. In conclusion, preexposure to an anti-inflammatory drug improved rats' resilience to long-term allodynia and pain-related manifestations. This suggests the immune system's involvement in the susceptibility to chronic pain and its comorbidities. Regarding mechanisms, Nrf2 and its inflammatory downstream effectors, as well as ATF-3, play a significant role, although potential contributions of other factors cannot be dismissed.
Collapse
Affiliation(s)
- Anh Cong Tuan Le
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Juliana Fiuza-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Margarida Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mariana Teixeira Sampaio
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andreia Texeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Hugo Leite-Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
5
|
Dogra A, Savitt AG, Ghebrehiwet B. C1q Binds to CD4+ T Cells and Inhibits the Release of Pro-Inflammatory Cytokines: Role in the Pathogenesis of Systemic Lupus Erythematosus. Int J Mol Sci 2025; 26:4468. [PMID: 40429614 DOI: 10.3390/ijms26104468] [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: 01/23/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
The association between C1q deficiency and the development of Systemic Lupus Erythematosus (SLE) is well established. Several studies have shown that deficiency in C1q is associated with failed apoptotic cleanup, leading to SLE progression. However, the magnitude of this correlation indicates that C1q may play a much more complex role in the development of lupus. This study provides further insight into the pathogenesis of SLE by investigating the consequences of the interaction between C1q and CD4+ T-cells in the breakdown of self-tolerance. Since the C1q/C1q receptor interaction is postulated to play a role, we first confirmed the presence of surface-expressed C1q and C1q receptors on CD4+ T-cells. Then, cell proliferation assays were performed in the presence and absence of purified C1q, gC1qR, and cC1qR. The supernatants of these cultures were used to determine the levels of immunoregulatory cytokines released. Our data confirm that increasing concentrations of C1q and gC1qR significantly inhibited cell proliferation. Furthermore, the CD4+ cells treated with either C1q or gC1qR secreted reduced inflammatory cytokines, such as IL-6 and TNF-alpha, compared to the untreated controls, suggesting that C1q deficiency facilitates the uncontrolled secretion of these critical cytokines, thus contributing to SLE. Although the role of pro-inflammatory cytokines in the induction of SLE is well documented, the mechanism by which C1q contributes to the disease is still a study in progress. Our data demonstrate that the interaction between C1q and its receptors on CD4+ T cells plays a critical role in the suppression of pro-inflammatory cytokines that cause tissue injury in SLE. Therefore, the C1q-C1qR axis may provide a rationally sound target for the design of novel therapeutic approaches for SLE treatment.
Collapse
Affiliation(s)
- Arushi Dogra
- Division of Rheumatology, Allergy and Clinical Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Anne G Savitt
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Berhane Ghebrehiwet
- Division of Rheumatology, Allergy and Clinical Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
6
|
Liu SCH, Chang K, Chen ML, Kuo MC, Wang TC, Wu RM. Oligodendrocyte-specific overexpression of human alpha-synuclein results in elevated MBP levels and inflammatory responses in TgM83 mice, mimicking the pathological features of multiple system atrophy. Acta Neuropathol Commun 2025; 13:94. [PMID: 40336104 PMCID: PMC12060544 DOI: 10.1186/s40478-025-02014-y] [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: 03/06/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025] Open
Abstract
Multiple system atrophy (MSA) is a neurodegenerative disorder characterized by parkinsonism, cerebellar dysfunction, and autonomic failure. Key pathological features of MSA include the formation of glial cytoplasmic inclusions (GCIs) in oligodendrocytes (OLs), myelin loss, and neuroinflammation. Although both inflammation and myelination are known to be critical in MSA, the roles of myelin proteins and their relationship with inflammation have often been overlooked. In this study, we injected AAV-Olig001 vectors carrying either human SNCA (AAV-hSNCA) or eGFP (AAV-eGFP) into the striatum of TgM83 transgenic mice, which express the A53T mutant form of human alpha-synuclein (αSyn), as well as into wild-type (WT) mice. We then assessed myelin protein expression and inflammatory responses. TgM83 mice injected with AAV-hSNCA exhibited demyelination, increased activation of microglia and astrocytes, and altered cytokine and chemokine profiles (including IL-1α, IL-10, IL-12(p40), CCL2, CCL3, CCL4, and CCL5), compared to both WT mice and TgM83 mice injected with AAV-eGFP. Interestingly, myelin basic protein (MBP) levels were significantly elevated around the injection site in TgM83 mice injected with AAV-hSNCA. Notably, we observed a positive correlation between MBP expression and inflammatory markers, as indicated by Iba1 and GFAP staining. These findings suggest that hSNCA overexpression is associated with increased MBP levels and enhanced inflammatory responses, implicating that MBP and myelination processes may play previously underappreciated roles in the pathogenesis of MSA.
Collapse
Affiliation(s)
- Sam Chi-Hao Liu
- Department of Neurology, National Taiwan University College of Medicine, No.1, Jen Ai Road section 1, Taipei City, Taiwan (R.O.C.)
- Department of Neurology, National Taiwan University Hospital, No.7, Chung Shan South Road, Taipei City, Taiwan (R.O.C.)
| | - Koping Chang
- Department of Pathology, National Taiwan University Hospital, No.7, Chung Shan South Road, Taipei City, Taiwan (R.O.C.)
| | - Meng-Ling Chen
- Department of Neurology, National Taiwan University College of Medicine, No.1, Jen Ai Road section 1, Taipei City, Taiwan (R.O.C.)
- Department of Neurology, National Taiwan University Hospital, No.7, Chung Shan South Road, Taipei City, Taiwan (R.O.C.)
| | - Ming-Che Kuo
- Department of Neurology, National Taiwan University College of Medicine, No.1, Jen Ai Road section 1, Taipei City, Taiwan (R.O.C.)
- Department of Medicine, National Taiwan University Cancer Center, No.57, Ln. 155, Keelung Road Section 3, Taipei City, Taiwan (R.O.C.)
| | - Teh-Cheng Wang
- Department of Neurology, National Taiwan University College of Medicine, No.1, Jen Ai Road section 1, Taipei City, Taiwan (R.O.C.)
| | - Ruey-Meei Wu
- Department of Neurology, National Taiwan University College of Medicine, No.1, Jen Ai Road section 1, Taipei City, Taiwan (R.O.C.).
- Department of Neurology, National Taiwan University Hospital, No.7, Chung Shan South Road, Taipei City, Taiwan (R.O.C.).
| |
Collapse
|
7
|
Tateishi A, Miyamoto T, Ohya M, Shiozawa T, Kanno H. Differences in lymphocyte subsets of spiral artery associated with impaired vascular remodeling in hypertensive disorders of pregnancy. BMC Pregnancy Childbirth 2025; 25:535. [PMID: 40329261 PMCID: PMC12054229 DOI: 10.1186/s12884-025-07653-6] [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: 11/23/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Hypertensive disorders of pregnancy (HDP) is assumed to be triggered by incomplete remodeling of the decidual spiral arteries. We examined the lymphocyte subsets and identified immune differences in the spiral arteries between HDP and normal pregnancies, and between remodeled and non-remodeled arteries such as the muscular artery and acute atherosis. One hundred seventy-three patients with HDP diagnosed at our hospital between 2008 and 2017 were included. Patients with a normal course of delivery and patients with premature birth after 34 weeks without pathological changes and abnormalities in the pregnancy course were included in the control group. The infiltrating lymphocytes were evaluated using immunohistochemistry. Acute atherosis was observed in 56 patients (32.4%) of all HDP patients. In patients that presented HDP with acute atherosis, the density of CD8+ T cells and CD4+ T cells was higher in the acute atherosis than those in the non-remodeled muscular artery. CD4+ T cells were more abundant in the non-remodeled muscular artery compared to the remodeled artery. In control patients, the density of CD4+ T cells was higher in the non-remodeled muscular artery than that in the remodeled artery; there was no difference in the density of CD56+ natural killer cells. There was no difference in the density of CD3+ T cells and CD56+ natural killer cells in the remodeled artery between patients presenting HDP with acute atherosis and control patients. These immune differences may cause changes in local cytokine balance around the spiral artery, contributing to the development of HDP.
Collapse
Affiliation(s)
- Ayako Tateishi
- Department of Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| | - Tsutomu Miyamoto
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390- 8621, Japan
| | - Maki Ohya
- Department of Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Tanri Shiozawa
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390- 8621, Japan
| | - Hiroyuki Kanno
- Department of Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| |
Collapse
|
8
|
Rodrigues A, Weber JI, Durães-Oliveira J, Moreno C, Ferla M, de Aires Pereira M, Valério-Bolas A, de Freitas BE, Nunes T, Antunes WT, Alexandre-Pires G, Pereira da Fonseca I, Santos-Gomes GM. Extracellular Vesicles Derived from Trypanosomatids: The Key to Decoding Host-Parasite Communication. Int J Mol Sci 2025; 26:4302. [PMID: 40362539 PMCID: PMC12072767 DOI: 10.3390/ijms26094302] [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: 03/18/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Trypanosomatids constitute a family of parasitic protozoa that cause significant human and veterinary diseases that are classified as neglected zoonotic diseases (NZDs). In a rapidly evolving world, these diseases have the potential to become a world health problem no longer solely associated with low-income countries. Therefore, the development of new strategies to control and restrain the dissemination of trypanosomatids is imperative. Extracellular vesicles (EVs) are a heterogeneous group of membrane-enclosed vesicles released by prokaryotic and eukaryotic cells. They can be found in diverse body fluids that carry biologically active molecules, including proteins, nucleic acids, lipids, and carbohydrates. EVs participate in cell-to-cell communication by delivering their cargo content to recipient cells. Thus, EVs play a role in regulating normal physiological processes, including immune surveillance and tissue repair, as well as being involved in pathological conditions, like cancer. In recent years, EVs have attracted significant attention from the scientific community, mainly due to their immune regulatory properties. Therefore, this review examines the role played by trypanosomatid-derived EVs in leishmaniases and trypanosomiasis, highlighting their biological role in host-parasite communication and exploring their potential future applications in controlling NZDs, especially those caused by trypanosomatids.
Collapse
Grants
- EXPL/CVT-CVT/0175/2021 (DOI 10.54499/EXPL/CVT-CVT/0175/2021) FCT-Foundation for Science and Technology, I.P.
- FPTDC/CVT-CVT/0228/2020 (DOI 10.54499/PTDC/CVT-CVT/0228/2020) FCT-Foundation for Science and Technology, I.P.
- CIISA, UIDB/00276/2020 FCT-Foundation for Science and Technology, I.P.
- Al4Animals, LA/P/0059/2020 FCT-Foundation for Science and Technology, I.P.
- CERNAS, UIDB/00681/2020 Foundation for Science and Technology, I.P.
- GHTM, UID/04413/2020 Foundation for Science and Technology, I.P.
- LA-REAL, LA/P/0117/2020) Foundation for Science and Technology, I.P.
- CEECIND/CP1725/CT0023 (10.54499/2022.00499.CEECIND/CP1725/CT0023) FCT-Foundation for Science and Technology, I.P.
- 2022.13899.BD FCT-Foundation for Science and Technology, I.P.
Collapse
Affiliation(s)
- Armanda Rodrigues
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
| | - Juliana Inês Weber
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
| | - João Durães-Oliveira
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
| | - Cláudia Moreno
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
| | - Micheli Ferla
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
| | - Maria de Aires Pereira
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
- CERNAS-IPV Research Centre, Instituto Politécnico de Viseu, Campus Politécnico, Repeses, 3504-510 Viseu, Portugal
| | - Ana Valério-Bolas
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
| | - Bruna Eugênia de Freitas
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
- Instituto de Ciências Biológicas, ICB, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Telmo Nunes
- Microscopy Center, Faculty of Sciences, University of Lisbon-FCUL-BioISI Ce3CE, 1749-016 Lisboa, Portugal;
| | - Wilson T. Antunes
- Instituto Universitário Militar (IUM), Centro de Investigação, Desenvolvimento e Inovação da Academia Militar (CINAMIL), Unidade Militar Laboratorial de Defesa Biológica e Química (UMLDBQ), 1849-012 Lisboa, Portugal;
| | - Graça Alexandre-Pires
- CIISA, Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1649-004 Lisbon, Portugal; (G.A.-P.); (I.P.d.F.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1200-771 Lisbon, Portugal
| | - Isabel Pereira da Fonseca
- CIISA, Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1649-004 Lisbon, Portugal; (G.A.-P.); (I.P.d.F.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1200-771 Lisbon, Portugal
| | - Gabriela M. Santos-Gomes
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisboa, Portugal; (J.I.W.); (J.D.-O.); (C.M.); (M.F.); (M.d.A.P.); (A.V.-B.); (B.E.d.F.); (G.M.S.-G.)
| |
Collapse
|
9
|
de Oliveira IM, Chaves MM. The NLRP3 Inflammasome in inflammatory diseases: Cellular dynamics and role in granuloma formation. Cell Immunol 2025; 411-412:104961. [PMID: 40339528 DOI: 10.1016/j.cellimm.2025.104961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
The innate immune system recognizes pathogen-associated molecular patterns (PAMPs) and damage associated molecular patterns (DAMPs) through pattern recognition receptors (PRRs). Inflammasomes, cytoplasmic protein complexes, are activated in response to PAMPs and DAMPs, leading to the release of inflammatory cytokines such as IL-1β and IL-18. NLRP3 inflammasome is one of the best characterized inflammasomes and recently its activation has been associated with granuloma formation, structures that aggregate immune cells in response to infections, such as those caused by bacteria, fungi and parasites, and autoinflammatory diseases, such as sarcoidosis. Activation of NLRP3 inflammasomes in macrophages induces the release of cytokines that recruit immune cells, such as monocytes and lymphocytes, to the site of infection. Neutrophils, monocytes, T and B lymphocytes are important in the formation and maintenance of granulomas. Although NLRP3 plays a key role in the immune response, cell recruitment and granuloma formation, many aspects of its function in different cell types remain to be elucidated. In this review, we aim to outline the NLRP3 inflammasome not only as a protein complex that aids innate immune cells in combating intracellular pathogens but also as a platform with broader implications in orchestrating immune responses. This underexplored aspect of the NLRP3 inflammasome presents a novel perspective on its involvement in immunity. Thus, we review the current understanding of the role of the NLRP3 inflammasome in immune cell infiltration and its significance in the organization and formation of granulomas in inflammatory diseases.
Collapse
Affiliation(s)
- Isadora M de Oliveira
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mariana M Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil; Bio-Manguinhos, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
10
|
Ma T, Zhang T, Miao F, Liu J, Zhu Q, Chen Z, Tai Z, He Z. Alopecia Areata: Pathogenesis, Diagnosis, and Therapies. MedComm (Beijing) 2025; 6:e70182. [PMID: 40260013 PMCID: PMC12010142 DOI: 10.1002/mco2.70182] [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: 10/09/2024] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Alopecia areata (AA) is a complex, chronic inflammatory skin disorder characterized by unpredictable, nonscarring hair loss, affecting millions worldwide. Its pathogenesis remains poorly understood, driven by intricate interactions among immune dysregulation, genetic predisposition, and environmental triggers. Despite significant advances in identifying these contributing factors, substantial gaps persist in our understanding of the full spectrum of AA's molecular mechanisms and in the development of effective therapeutic approaches. This review aims to comprehensively explore the immunological, genetic, epigenetic, and environmental factors underlying AA, with a focus on immune-mediated mechanisms. We also evaluate diagnostic approaches and recent advancements in assessing disease severity. Furthermore, the review discusses evolving therapeutic options, including traditional therapies, biologics, small-molecule agents, and emerging treatments. The academic value of this work lies in its synthesis of current knowledge on the multifaceted nature of AA, providing insights for future research and clinical practice. By elucidating the interconnected factors underlying AA, this review seeks to advance both understanding and management of this prevalent, clinically challenging disorder.
Collapse
Affiliation(s)
- Tianyou Ma
- Department of PharmacyLonghua Hospital of Shanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Tingrui Zhang
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Fengze Miao
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Jun Liu
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Quangang Zhu
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Zhongjian Chen
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Zongguang Tai
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Zhigao He
- Department of PharmacyLonghua Hospital of Shanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
11
|
Browne DJ, Crooks P, Smith C, Doolan DL. Differential reactivity of SARS-CoV-2 S-protein T-cell epitopes in vaccinated versus naturally infected individuals. Clin Transl Immunology 2025; 14:e70031. [PMID: 40342296 PMCID: PMC12056234 DOI: 10.1002/cti2.70031] [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: 11/18/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 05/11/2025] Open
Abstract
Objectives Vaccine-induced protective immunity against SARS-CoV-2 has proved difficult to sustain. Robust T-cell responses are thought to play an important role, but T-cell responses against the SARS-CoV-2 spike protein (S-protein), the core vaccine antigen, following vaccination or natural infection are incompletely understood. Methods Herein, the reactivity of 170 putative SARS-CoV-2 S-protein CD8+ and CD4+ T-cell peptide epitopes in the same individuals prior to vaccination, after COVID-19 vaccination, and again following subsequent natural infection was assayed using a high-throughput reverse transcription-quantitative PCR (HTS-RT-qPCR) assay. Results The profile of immunoreactive SARS-CoV-2 S-protein epitopes differed between vaccination and natural infection. Vaccine-induced immunoreactive epitopes were localised primarily into two extra-domanial regions. In contrast, epitopes recognised following natural infection were spread across the antigen. Furthermore, T-cell epitopes in naïve individuals were primarily recognised in association with HLA-A, while natural infection shifted epitope associations towards HLA-B, particularly the B7 supertype. Conclusion This study provides insight into T-cell responses against the SARS-CoV-2 S-protein following vaccination and subsequent natural infection.
Collapse
Affiliation(s)
- Daniel J Browne
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQLDAustralia
| | - Pauline Crooks
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of ImmunologyQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of ImmunologyQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
- Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQLDAustralia
- Institute for Molecular BioscienceThe University of QueenslandSt LuciaQLDAustralia
| |
Collapse
|
12
|
Huang YE, Zhou S, Chen S, Chen J, Zhou X, Hou F, Liu H, Yuan M, Jiang W. Mutational signature-based biomarker to predict the response of immune checkpoint inhibitors therapy in cancers. Int J Biol Macromol 2025; 308:142585. [PMID: 40154701 DOI: 10.1016/j.ijbiomac.2025.142585] [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: 07/18/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Patients have a limited response rate to immune checkpoint inhibitors (ICIs) therapy. Although several biomarkers have been proposed, their ability to accurately predict the response to ICIs therapy remains unsatisfactory. In addition, mutational signatures were validated to be associated with ICIs therapy. Therefore, we developed a mutational signature-based biomarker (MS-bio) to predict the response to ICIs therapy. Based on differentially mutated genes, we extracted six mutational signatures (single-base substitution (SBS)-A, SBS-B, SBS-C, SBS-D, double-base substitution (DBS)-A, and DBS-B) as MS-bio, and constructed a random forest (RF) model to predict the response. Internal and external validations consistently demonstrated the excellent predictive capability of MS-bio, with an accuracy reaching up to 0.82. Moreover, MS-bio exhibited superior performance compared to existing biomarkers. To further validate the accuracy of MS-bio, we explored its performance in The Cancer Genome Atlas (TCGA) cohort and found that the predicted responders were immunologically "hot". Finally, we found that SBS-C had the highest importance in prediction and was related to T cell differentiation. Overall, here we introduced MS-bio as a novel biomarker for accurately predicting the response to ICIs therapy, thereby contributing to the advancement of precision medicine.
Collapse
Affiliation(s)
- Yu-E Huang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Guizhou Institute of Precision Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Shunheng Zhou
- School of Computer Sciences, University of South China, Hengyang 421001, China
| | - Sina Chen
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Jiahao Chen
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Xu Zhou
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Fei Hou
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Haizhou Liu
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Mengqin Yuan
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Wei Jiang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
13
|
Aiman S, Ahmad A, Malik A, Chen R, Hanif MF, Khan AA, Ansari MA, Farrukh S, Xu G, Shahab M, Huang K. Whole proteome-integrated and vaccinomics-based next generation mRNA vaccine design against Pseudomonas aeruginosa-A hierarchical subtractive proteomics approach. Int J Biol Macromol 2025; 309:142627. [PMID: 40174835 DOI: 10.1016/j.ijbiomac.2025.142627] [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: 10/09/2024] [Revised: 03/09/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a multidrug-resistant opportunistic pathogen responsible for chronic obstructive pulmonary disease (COPD), cystic fibrosis, and ventilator-associated pneumonia (VAP), leading to cancer. Developing an efficacious vaccine remains the most promising strategy for combating P. aeruginosa infections. In this study, we employed an advanced in silico strategy to design a highly efficient and stable mRNA vaccine using immunoinformatics tools. Whole proteome data were utilized to identify highly immunogenic vaccine candidates using subtractive proteomics. Three extracellular proteins were prioritized for T- and linear B-cell epitope prediction. Beta-definsin protein sequence was incorporated as an adjuvant at the N-terminus of the construct. A total of 3 CTL, 3 HTL, and 3 linear B cell highly immunogenic epitopes were combined using specific linkers to design this multi-peptide construct. The 5' and 3' UTR sequences, Kozak sequence with a stop codon, and signal peptides followed by a poly-A tail were incorporated into the above vaccine construct to create our final mRNA vaccine. The vaccines exhibited antigenicity scores >0.88, ensuring high antigenicity with no allergenic or toxic. Physiochemical properties analysis revealed high solubility and thermostability. Three-dimensional structural analysis determined high-quality structures. Vaccine-receptor docking and molecular dynamic simulations demonstrated strong molecular interactions, stable binding affinities, dynamic nature, and structural stability of this vaccine, with significant immunogenic responses of the immune system against the vaccine. The immunological simulation indicates successful cellular and humoral immune responses to defend against P. aeruginosa infection. Validation of the study outcomes necessitates both experimental and clinical testing.
Collapse
Affiliation(s)
- Sara Aiman
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Liaobu Hospital of Dongguan City, Dongguan, China
| | - Abbas Ahmad
- Department of Biotechnology, Abdul Wali Khan University, Mardan, Pakistan
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Rui Chen
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Muhammad Farhan Hanif
- Department of Energy and Resource Engineering, College of Engineering, Peking University, Beijing 100871, China.
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Mushtaq Ahmed Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | | | - Guangxian Xu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
| | - Muhammad Shahab
- State key laboratories of chemical Resources Engineering Beijing University of Chemical Technology, Beijing 100029, China.
| | - Kaisong Huang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Liaobu Hospital of Dongguan City, Dongguan, China.
| |
Collapse
|
14
|
Chaudhari R, Dasgupta M, Kodgire P. Unravelling the Impact of Outer Membrane Protein, OmpA, From S. Typhimurium on Aberrant AID Expression and IgM to IgA Class Switching in Human B-Cells. Immunology 2025. [PMID: 40300848 DOI: 10.1111/imm.13938] [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: 08/13/2024] [Revised: 02/12/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025] Open
Abstract
Salmonella enterica serovar Typhimurium is a Gram-negative bacterium that causes gastrointestinal infection and poses significant public health risks worldwide. This study aims to explore how S. Typhimurium manipulates B-cell function through outer membrane protein A (OmpA). We investigate the effect of OmpA on Raji human B-cells, leading to the induction of activation-induced cytidine deaminase (AID) protein, which plays an important role in generating antibody diversity in B-cells, via initiating the process of somatic hypermutation (SHM) and class switch recombination (CSR). Our key findings demonstrate that OmpA is crucial for inducing aberrant AID expression in B-cells, leading to increased CSR. Interestingly, the increased AID expression was likely due to overexpression of cMYC, an activator for AID expression. Not only was the expression of cMYC elevated, but its occupancy on the aicda locus was raised. Furthermore, increased AID expression induced CSR events, specifically switching to IgA. In summary, our study suggests that OmpA plays a potential role in modulating B-cell regulation and controlling the adaptive immune system. These functional attributes of OmpA implicate its potential as a therapeutic target for combating S. Typhimurium pathogenesis.
Collapse
Affiliation(s)
- Rahul Chaudhari
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, Indore, India
| | - Mallar Dasgupta
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, Indore, India
| | - Prashant Kodgire
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, Indore, India
| |
Collapse
|
15
|
Otaegui J, Sultan D, Heo GS, Liu Y. Positron Emission Tomography Imaging of the Adaptive Immune System in Cardiovascular Diseases. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:209-224. [PMID: 40313531 PMCID: PMC12042138 DOI: 10.1021/cbmi.4c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 05/03/2025]
Abstract
Cardiovascular diseases are the leading cause of death around the globe. In recent years, a crucial role of the immune system has been acknowledged in cardiac disease progression, opening the door for immunomodulatory therapies. To this ongoing change of paradigm, positron emission tomography (PET) imaging of the immune system has become a remarkable tool to reveal immune cell trafficking and monitor disease progression and treatment response. Currently, PET imaging of the immune system in cardiovascular disease mainly focuses on the innate immune system such as macrophages, while the immune cells of the adaptive immune system including B and T cells are less studied. This can be ascribed to the lack of radiotracers specifically binding to B and T cell biomarkers compatible with PET imaging within the cardiovascular system. In this review, we summarize current knowledge about the role of the adaptive immune system (e.g., B and T cells) in major cardiovascular diseases and introduce key biomarkers for specific targeting of these immune cells and their subpopulations. Finally, we present available radiotracers for these biomarkers and propose a pathway for developing probes or optimizing those already used in other fields (e.g., oncology) to make them compatible with the cardiovascular system.
Collapse
Affiliation(s)
- Jaume
Ramon Otaegui
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Deborah Sultan
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Gyu Seong Heo
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Yongjian Liu
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| |
Collapse
|
16
|
Lian Z, Luo Y, Li Y, Gao Y, Xiong X, Gu L. CD4 + T cells in ischemic stroke: effects and therapeutic targets. Front Immunol 2025; 16:1512634. [PMID: 40352928 PMCID: PMC12061934 DOI: 10.3389/fimmu.2025.1512634] [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: 10/18/2024] [Accepted: 03/27/2025] [Indexed: 05/14/2025] Open
Abstract
Ischemic stroke (IS) is a significant contributor to disability and death worldwide, with limited treatments beyond early intervention. The importance of CD4+ T cells in the advancement of IS has been highlighted by recent studies, providing new insights into immunomodulatory strategies. This review describes the spatiotemporal dynamics of CD4+ T cells and their subsets at different stages of IS. The signaling pathways activated by IS regulate the distribution of CD4+ T cells and their subsets, which further influences the inflammatory response and disease progression. In the acute and subacute stages, CD4+ T cells exacerbate neuronal damage. In contrast, CD4+ T cells, which are predominantly composed of Treg cells (Tregs), promote tissue repair and neurological recovery in the chronic stage. In light of recent findings that challenge traditional views, we analyze the underlying mechanisms and potential explanations for these discrepancies. In addition, we summarize the potential of targeting CD4+ T cells as a therapeutic strategy for IS. Although no drugs specifically targeting CD4+ T cells have been developed, certain drugs that modulate CD4+ T cells show potential for IS treatment. Moreover, multitarget drugs integrated with nanomaterials are currently undergoing preclinical investigation. We further explore the challenges in the clinical translation of CD4+ T-cell-targeted therapies and discuss potential strategies to address these challenges. In conclusion, a deeper comprehension of the complex effects of CD4+ T cells and their subsets on IS will contribute to disease management and drug development, thereby improving the quality of life for IS patients.
Collapse
Affiliation(s)
- Zhengqi Lian
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ying Luo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yikun Gao
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Telec M, Frydrychowicz M, Kazmierski R, Wojtasz I, Dworacki G, Kozubski W, Łukasik M. Circulating CD4+, CD8+, and double-negative T cells in ischemic stroke and stroke-associated infection: a prospective case-control study. Front Cell Neurosci 2025; 19:1547905. [PMID: 40342517 PMCID: PMC12058799 DOI: 10.3389/fncel.2025.1547905] [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: 12/18/2024] [Accepted: 02/24/2025] [Indexed: 05/11/2025] Open
Abstract
Introduction Adaptive immunity after a stroke results in a shift of T cells between compartments, leading to peripheral lymphopenia and an increased number of T cells within the brain lesion. Stroke-associated infection (SAI) presents a clinically significant challenge in stroke units. The role of T-cell subsets in the post-stroke immune response and in SAI remains unclear. Thus, we aimed to observe the quantitative changes of circulating CD4+, CD8+, double-negative T cells, and the CD4+/CD8+ ratio in stroke and SAI. Methods We prospectively assessed circulating CD4+, CD8+, and double-negative T cells using flow cytometry in 52 patients on days 1, 3, 10, and 90 after ischemic stroke. We compared the results to those obtained from age-, sex-, and vascular risk factor-matched controls. We analyzed lymphocyte parameters in relation to clinical outcome, SAI, infarct lesion volume, and risk factor burden. Results There were no differences in the studied parameters between stroke patients and controls, as well as between subjects with and without SAI. A higher percentage of CD4+ T cells and a higher CD4+/CD8+ ratio correlated with better clinical status in the acute and subacute phases, while CD8+ T cells showed the opposite correlation. The percentage of CD8+ T cells positively correlated with CRP levels during the acute and subacute phases of stroke, as well as in the control group. A negative correlation was noted between the percentage of CD4+ T cells on D1 and the serum CRP level on D10 after stroke. Similarly, the CD4+/CD8+ ratio on D1 negatively correlated with CRP on D1, D3, and D10. In patients with a history of hypertension (HT), there was a higher percentage of CD8+ T cells and a lower percentage of CD4+ T cells in the acute phase of stroke than those without HT.
Collapse
Affiliation(s)
- Magdalena Telec
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Radosław Kazmierski
- Department of Neurology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| | - Izabela Wojtasz
- Department of Neurology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| | - Grzegorz Dworacki
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maria Łukasik
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
18
|
Galsuren J, Dambadarjaa D, Tighe RM, Gray GC, Zhang J. Particulate Matter Exposure and Viral Infections: Relevance to Highly Polluted Settings such as Ulaanbaatar, Mongolia. Curr Environ Health Rep 2025; 12:22. [PMID: 40268823 DOI: 10.1007/s40572-025-00484-9] [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] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
PURPOSE OF REVIEW Particulate matter (PM), a ubiquitous significant component of the ambient air pollution mixture, significantly contributes to increased global risk for chronic cardiopulmonary diseases, acute hospitalizations, and deaths. One of the causes of this increased risk is because PM exposure increases the incidence and severity of respiratory infections. The respiratory system is particularly vulnerable to air pollution and its impact on infection as it is a key site for exposure both to inhaled pollutants and infectious microbes or viruses. This review examines the current understanding of how PM affects antiviral host defense responses and possible underlying mechanisms. RECENT FINDINGS While numerous studies have associated adverse health outcomes with combined or sequential exposure to inhaled pollutants and viruses, defining causal relationships and mechanisms remains limited. Particularly limited, are contemporary data focuses on low- and middle-income countries, including heavily polluted regions such as Ulaanbaatar, Mongolia. This manuscript focuses on how (1) PM, serving as a carrier for viruses, enhances the transmission of viruses; (2) PM impairs immune defense to viruses; and (3) PM impacts epithelial cell functions to exacerbate viral infections. Given the significant public health hazards on PM, particularly in heavily polluted regions such as Southeast Asia, Middle East and Africa, it is critical to define specific mechanisms of PM on respiratory infection and how their impact may differ in these highly polluted regions. Ultimately, this could devise future public health measures and interventions to limit this substantial public health risk.
Collapse
Affiliation(s)
- Jargalsaikhan Galsuren
- School of Public Health, Mongolian National University of Medical Sciences, Ulaanbaatar, 14210, Mongolia
| | - Davaalkham Dambadarjaa
- School of Public Health, Mongolian National University of Medical Sciences, Ulaanbaatar, 14210, Mongolia
| | - Robert M Tighe
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Duke University, Durham, NC, 27710, USA
| | - Gregory C Gray
- Department of Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Junfeng Zhang
- Duke Nicholas School of the Environment, Durham, NC, 27705, USA.
| |
Collapse
|
19
|
Kline EM, Jernigan JE, Scharer CD, Maurer J, Hicks SL, Herrick MK, Wallings RL, Kelly SD, Chang J, Menees KB, McFarland NR, Boss JM, Tansey MG, Joers V. MHCII reduction is insufficient to protect mice from alpha-synuclein-induced degeneration and the Parkinson's HLA locus exhibits epigenetic regulation. Sci Rep 2025; 15:13705. [PMID: 40258905 PMCID: PMC12012047 DOI: 10.1038/s41598-025-95679-3] [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: 09/02/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Major histocompatibility complex class II (MHCII) molecules are antigen presentation proteins and increased in post-mortem Parkinson's disease (PD) brain. Attempts to decrease MHCII expression have led to neuroprotection in PD mouse models. Our group reported that a single nucleotide polymorphism (SNP) at rs3129882 in the MHCII gene Human Leukocyte Antigen (HLA) DRA is associated with increased MHCII transcripts and surface protein and increased risk for late-onset idiopathic PD. We therefore hypothesized that decreased MHCII may mitigate dopaminergic degeneration. During an ongoing α-synuclein lesion, mice with MHCII reduction in systemic and brain innate immune cells (LysMCre + I-Abfl/fl or CRE+) displayed brain T cell repertoire shifts and greater preservation of the dopaminergic phenotype in nigrostriatal terminals. Next, we investigated a human cohort to characterize the immunophenotype of subjects with and without the high-risk GG genotype at the rs3129882 SNP. We confirmed that the high-risk GG genotype is associated with peripheral changes in MHCII inducibility, frequency of CD4 + T cells, and differentially accessible chromatin regions within the MHCII locus. Although our mouse studies indicate that myeloid MHCII reduction coinciding with an intact adaptive immune system is insufficient to fully protect dopamine neurons from α-synuclein-induced degeneration, our data are consistent with the overwhelming evidence implicating antigen presentation in PD pathophysiology.
Collapse
Affiliation(s)
- Elizabeth M Kline
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Janna E Jernigan
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida Health, Gainesville, FL, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeffrey Maurer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sakeenah L Hicks
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mary K Herrick
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida Health, Gainesville, FL, USA
| | - Rebecca L Wallings
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida Health, Gainesville, FL, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sean D Kelly
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jianjun Chang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelly B Menees
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida Health, Gainesville, FL, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nikolaus R McFarland
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida Health, Gainesville, FL, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| | - Valerie Joers
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA.
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida Health, Gainesville, FL, USA.
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
20
|
Lam L, Pietris J, Quigley C, Davis G, Slattery J, Selva D. Bilateral sequential bacterial dacryoadenitis with abscess- a case report. BMC Ophthalmol 2025; 25:227. [PMID: 40251557 PMCID: PMC12007365 DOI: 10.1186/s12886-025-04063-2] [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: 12/05/2024] [Accepted: 04/10/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Acute dacryoadenitis is characterized by inflammation of the lacrimal gland, and is typically viral. Bacterial dacryoadenitis is rare, with Staphylococcus and Streptococcus species being the most common pathogens. The bilateral sequential onset of dacryoadenitis has not been described in the literature to date. CASE PRESENTATION A 54-year-old immunocompetent female presented to the emergency department with a 1-week history of left-sided periorbital swelling, erythema, and pain. Orbital computed tomography demonstrated significant soft tissue thickening and swelling surrounding the left orbit and a lacrimal gland abscess. Culture swabs were negative for microorganisms. Despite initial management with intravenous antibiotics and surgical drainage, the condition recurred post-discharge. Three years later, the patient experienced similar symptoms in the right eye. Cultures were repeatedly negative for microorganisms. Despite courses of intravenous antibiotics and surgical drainage, the patient experienced recurrences of abscesses in the right eye, highlighting the challenges in managing this rare condition. CONCLUSIONS We describe a unique case of bilateral sequential bacterial dacryoadenitis with abscess formation. This case highlights the need for prompt diagnosis and management, including surgical intervention, to prevent complications and recurrences.
Collapse
Affiliation(s)
- Lydia Lam
- The University of Adelaide, Adelaide Medical School, Adelaide, SA, 5005, Australia.
| | - James Pietris
- Department of Ophthalmology and South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Clare Quigley
- Department of Ophthalmology and South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Garry Davis
- Department of Ophthalmology and South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - James Slattery
- Department of Ophthalmology and South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Dinesh Selva
- Department of Ophthalmology and South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
21
|
Gong H, Nie D, Li Z. The crosstalk between broad epigenetic modification and T cell metabolism within tumor microenvironment. Int Immunopharmacol 2025; 152:114410. [PMID: 40068521 DOI: 10.1016/j.intimp.2025.114410] [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: 11/17/2024] [Revised: 03/01/2025] [Accepted: 03/02/2025] [Indexed: 03/24/2025]
Abstract
T cells play an important role in adaptive immune responses, providing antigen specificity for pathogen and tumor recognition. Recent studies have elucidated the complex interplay between T cell metabolism and broad epigenetic modifications in response to tumors, occurring at transcriptional, post-transcriptional, and post-translational levels. At the transcriptional level, gene expression is regulated through mechanisms such as DNA methylation, chromatin remodeling, and transcription factor activity. Post-transcriptionally, gene expression is further modulated by non-coding RNAs and RNA modifications, an area of increasing research interest. In addition, histone proteins are primarily regulated by well-established post-translational modifications (PTMs), including acetylation and methylation. Novel PTMs such as succinylation, glycosylation, glutamylation, and lactylation add complexity to the regulation and warrant further investigation. At present, the interaction between CD8+ T cell metabolism and epigenetic modifications in response to malignancies has been reported extensively. However, the interplay in CD4+ T cells remains less understood. In this review, we introduce the differentiation trajectories of T cells and critically evaluate existing interplay between metabolic activity and epigenetic modifications influences the functional dynamics in both CD8+ and CD4+ T cells, offering promising avenues for the development of novel cancer immunotherapies.
Collapse
Affiliation(s)
- Han Gong
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Dan Nie
- Department of Obstetrics and Gynecology, The affiliated hospital of Southwest Medical University, Luzhou 646000, People's Republic of China
| | - Zhengyu Li
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, People's Republic of China.
| |
Collapse
|
22
|
Wang JS, Schellenberg SJ, Demeros A, Lin AY. Exosomes in review: A new frontier in CAR-T cell therapies. Neoplasia 2025; 62:101147. [PMID: 40037165 PMCID: PMC11923832 DOI: 10.1016/j.neo.2025.101147] [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: 01/28/2025] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
Exosomes are extracellular vehicles that facilitate intra-cellular communication via transport of critical proteins and genetic material. Every exosome is intrinsically reflective of the cell from which it was derived and can even mimic effector functions of their parent cells. In recent years, with the success of CAR-T therapies, there has been growing interest in characterizing exosomes derived from CAR-T cells. CAR exosomes contain the same cytotoxic granules as their parent cells and have demonstrated significant anti-tumor activity in vitro and in animal models. Moreover, infusion of CAR exosomes in animal models did not generate cytokine release syndrome. Conversely, there are also novel bispecific antibodies which target tumor-derived exosomes in hopes of derailing immunosuppressive pathways mediated by exosomes produced from malignant cells. The two most promising examples include (a) BsE CD73 x EpCAM which binds and inhibits exosomal CD73 to suppress production of immunosuppressant adenosine and (b) BsE CD3 x PD-L1 which targets exosomal PD-L1 within the tumor microenvironment to guide cytotoxic T-cells towards tumor cells. As our understanding of exosome biology continues to evolve, opportunities for advances in cellular therapies will grow in tandem.
Collapse
Affiliation(s)
- John S Wang
- Northwestern University, Feinberg School of Medicine, Department of Medicine, Chicago, IL, USA
| | - Samuel J Schellenberg
- Northwestern University, Feinberg School of Medicine, Department of Medicine, Chicago, IL, USA
| | | | - Adam Y Lin
- Northwestern University, Feinberg School of Medicine, Department of Medicine, Division of Oncology, Chicago, IL, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| |
Collapse
|
23
|
Black C, Harvey SB, Holladay SD, Howerth E, Hogan RJ, Bergeron HC, Gogal RM. Revisiting tribromoethanol as a safe and effective murine anesthetic for veterinary clinical and biomedical research. Vet Immunol Immunopathol 2025; 282:110909. [PMID: 40024017 DOI: 10.1016/j.vetimm.2025.110909] [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: 01/02/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
Mus musculus, the house mouse, is the most widely used mammalian model in biomedical research. Mice frequently undergo injectable anesthesia for numerous research procedures, with the most common anesthetic protocol being ketamine-xylazine (K/X). 2,2,2-Tribromoethanol (TBE), a non-regulated chemical, is also used, but has been linked to peritonitis. The focus of this study was to directly compare these two anesthetic protocols by evaluating induction rates, recovery times, organ weight data, and immune endpoints. Forty-five CD-1 female (8-10 week-old) mice were divided into three experiments. Two anesthetic events were performed 2 weeks apart. For each experiment, mice received an intraperitoneal (IP) injection of sterile phosphate buffered saline, (PBS; n = 3 mice), an IP injection of K/X (n = 6 mice), or an IP injection of sterile TBE (n = 6 mice). In a separate third anesthetic event (n = 5 mice/treatment), post-treatment peripheral blood and peritoneal lavage samples were collected for a 9-plex cytokine analysis. Mice were euthanized 2 weeks after the last anesthetic event. Induction rates were non-significantly but numerically more rapid with TBE as compared to K/X, at 2.7 ± 0.6 min and 4.0 ± 0.7 min, respectively. TBE mice had significantly more rapid recovery time (∼25 min) compared to K/X (∼50 min), which also had 50 % anesthetic mortalities. Organ weight ratios, immune phenotype, cytology, serum and peritoneal lavage cytokine levels, and histopathology were unremarkable. TBE performed better and more safely as a murine anesthetic for light anesthesia compared to K/X based on recovery times, no mortalities, and an absence of local and systemic inflammation.
Collapse
Affiliation(s)
- Catherine Black
- Department of Population Health, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States; Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States
| | - Stephen B Harvey
- Department of Population Health, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States
| | - Steven D Holladay
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States
| | - Elizabeth Howerth
- Department of Pathology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States
| | - Robert Jeff Hogan
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States
| | - Harrison C Bergeron
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States
| | - Robert M Gogal
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602, United States.
| |
Collapse
|
24
|
Rendell M. Pharmacotherapy of type 1 diabetes - part 2 Today. Expert Opin Pharmacother 2025; 26:719-730. [PMID: 40082213 DOI: 10.1080/14656566.2025.2479598] [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: 12/07/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION In the 100 years since isolation and administration of animal insulin to sustain life in Type 1 diabetes, there has been increasing progress in the administration of exogenous insulin to lower glucose levels. AREAS COVERED We reviewed using standard search engines and PubMed present-day techniques of management of type 1 diabetes. EXPERT OPINION Long-acting insulin formulations have been developed to maintain basal glucose levels in the normal range, while rapid acting insulins have been synthesized to address the sharp rise in glucose levels after a meal. Insulin pumps administer insulin continuously subcutaneously guided by continuous glucose monitoring systems. These almost closed loop systems achieve near normal glucose levels other than at meal times where the rapid glucose rise and then fall pose a significant challenge due to the extended duration of subcutaneous insulin depots. Implanted insulin pumps with intraperitoneal delivery may eventually permit improved post meal glucose control. Type 1 diabetes has now been redefined as an autoimmune disease which may be diagnosed purely from the presence of anti-beta cell antibodies with no abnormality of glucose levels. The future will see an intensification of efforts to combat the immune process which destroys beta cells.
Collapse
Affiliation(s)
- Marc Rendell
- The Association of Diabetes Investigators, Omaha, NE, USA
- The Rose Salter Medica Research Foundation, Newport Coast, CA, USA
| |
Collapse
|
25
|
Salloom RJ, Sahtout DZ, Ahmad IM, Abdalla MY. Synergistic effects of HO-1 inhibition and chemotherapy on tumor proliferation and immune infiltration: An in vitro and in vivo approach to enhancing prostate cancer treatment. Transl Oncol 2025; 54:102339. [PMID: 40037158 PMCID: PMC11925535 DOI: 10.1016/j.tranon.2025.102339] [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: 09/09/2024] [Revised: 01/24/2025] [Accepted: 02/27/2025] [Indexed: 03/06/2025] Open
Abstract
Prostate cancer (PC) remains a leading cause of morbidity and mortality among men worldwide, highlighting the need for novel therapeutic strategies. Our study investigates the therapeutic potential of targeting the heme degradation pathway through heme oxygenase-1 (HO-1) inhibition in PC. Using both in vitro and in vivo models, we explored the effects of combining HO-1 inhibition with chemotherapy, represented by docetaxel (Doc), on tumor growth and immune infiltration. In vitro experiments demonstrated that HO-1 inhibition, as well as HO-1 knockout (KO), significantly reduced tumor cell proliferation and enhanced chemosensitivity in RM-1 cells. Additionally, U937 cells co-cultured with HO-1 KO cells shifted cell polarization toward an M1 phenotype. In vivo, the combined treatment of the HO-1 inhibitor, tin protoporphyrin (SnPP), with Doc significantly enhanced anti-tumor efficacy in mouse models compared to chemotherapy or SnPP alone. This combination therapy not only reduced Ki67 expression and increased CC3 expression in tumor tissues but also shifted macrophage polarization toward an M1 phenotype and enhanced CD4+ and CD8+ T cells infiltration, indicating an augmented immune response. Further investigation using macrophage-specific HO-1 knockout mice revealed a direct role of HO-1 inhibition in driving macrophage polarization, confirming its involvement in promoting the shift toward an M1 phenotype. Although this response was significant, it was more robust with systemic HO-1 inhibition. Our findings indicate that HO-1 inhibition can potentiate the effects of chemotherapy, offering a promising avenue for improving PC treatment outcomes.
Collapse
Affiliation(s)
| | | | - Iman M Ahmad
- Department of Clinical, Diagnostics, and Therapeutic Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | | |
Collapse
|
26
|
Yoo E, Jo Y, Park J, Hong SW. Immune tolerance to foreign antigens in the intestine: mechanisms mediated by CD4+ T cells. BMB Rep 2025; 58:158-168. [PMID: 40176601 PMCID: PMC12041928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025] Open
Abstract
The immune system encounters a diverse array of antigens, both self and foreign, necessitating mechanisms to maintain tolerance and prevent harmful inflammatory responses. CD4+ T cells, crucial in orchestrating immune responses, play a critical role in mediating tolerance to both self and foreign antigens. While the mechanisms of CD4+ T cell-mediated tolerance to self-antigens are well-documented, the understanding of tolerance to foreign antigens, including those from commensal microbes and food, remains incomplete. This review discusses recent progress in the mechanisms underlying immune tolerance to foreign antigens, with a focus on the role of CD4+ T cells. We explore how inflammatory and tolerogenic CD4+ T cell subsets are developed and maintained. Moreover, we delve into the complexities of immune responses to commensal microbes and food antigens by reviewing recent findings, highlighting the immunological contexts that shape immune tolerance. Understanding these mechanisms enhances our comprehension of how immune tolerance is established and sustained, providing insights into potential therapeutic approaches for managing chronic inflammatory diseases resulting from a loss of immune tolerance to foreign antigens. [BMB Reports 2025; 58(4): 158-168].
Collapse
Affiliation(s)
- Eunbi Yoo
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Yeleen Jo
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jooyoun Park
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Sung-Wook Hong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
27
|
Wang A, Li Z, Jiang Y, Chen M, Yu H, Li Z, Sun S, Bai G, Wang Q, Huang Y, Wang L. Prox1 Protein in Corneal Limbal Lymphatic Vessels Maintains Limbal Stem Cell Stemness and Regulates Corneal Injury Repair. Invest Ophthalmol Vis Sci 2025; 66:81. [PMID: 40298888 PMCID: PMC12045113 DOI: 10.1167/iovs.66.4.81] [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: 09/28/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Purpose The purpose of this study was to elucidate the role of the corneal lymphatic Prox1 gene in modulating limbal stem cell stemness and facilitating corneal injury repair. Methods The limbal Prox1 gene was knocked down by adeno-associated virus (AAV). The alkali burn model was induced in the naive group, the AAV-sham group, and the AAV-shProx1 group. Anterior segment photography, fluorescein sodium staining, and hematoxylin and eosin (H&E) staining were conducted immediately on days 1, 3, 7, 14, and 21 post-injury. Immunofluorescent (IF) staining was used to assess Ki67, ΔNp63, and K14. Additionally, seq-mRNA technology facilitated a comparative transcriptomic analysis between the AAV-sham and the AAV-shProx1 groups 7 days post-injury. Key regulated genes were verified by protein level. Furthermore, a co-culture model of lymphatic endothelial cells (LECs) and limbal stem cells (LSCs) was used to investigate the proliferation capacity and stemness expression of LSCs. Results Fluorescein sodium staining revealed that the epithelial defect area was significantly larger in the AAV-shProx1 group than in the AAV-sham group on days 1 and 3 post-injury (P < 0.05). Ki67, ΔNp63, and K14 expressions were consistently lower in the AAV-shProx1 group than in the AAV-sham group at distinct time points. Additionally, seq-mRNA results demonstrated that genes (Prox1 and Lyve1) were downregulated while inflammatory factors (Ccl2, Ccl7, IL16, IL1R, and TNFsf11) were upregulated in the AAV-shProx1 group compared with the AAV-sham group. When Prox1 was silenced in LECs, the proliferation and stemness of LSCs were markedly downregulated. Conclusions The Prox1 and Lyve1 proteins in lymphatic vessels served as pivotal regulated proteins in corneal injury repair. The draining role of lymphatic vessels during corneal injury was indispensable.
Collapse
Affiliation(s)
- An Wang
- Medical School of Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
| | - Zongyuan Li
- Medical School of Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
| | - Yilin Jiang
- Medical School of Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
| | - Mingxiong Chen
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Hanrui Yu
- Medical School of Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
| | - Zhao Li
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Shengshu Sun
- Medical School of Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
| | - Ge Bai
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Medical School of Jinzhou, Liaoning, China
| | - Qun Wang
- Medical School of Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
| | - Yifei Huang
- Medical School of Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
| | - Liqiang Wang
- Medical School of Chinese PLA General Hospital - 301 Hospital, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital - 301 Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
- Medical School of Jinzhou, Liaoning, China
| |
Collapse
|
28
|
Vruzhaj I, Gambirasi M, Busato D, Giacomin A, Toffoli G, Safa A. Gut Microbiota-Based Immunotherapy: Engineered Escherichia coli Nissle 1917 for Oral Delivery of Glypican-1 in Pancreatic Cancer. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:633. [PMID: 40282924 PMCID: PMC12028767 DOI: 10.3390/medicina61040633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: The administration of oral vaccines offers a potential strategy for cancer immunotherapy; yet, the development of effective platforms continues to pose a difficulty. This study examines Escherichia coli Nissle 1917 (EcN) as a microbial vector for the precise delivery of Glypican-1 (GPC1), a tumor-associated antigen significantly overexpressed in pancreatic ductal adenocarcinoma (PDAC).To evaluate the effectiveness of EcN as a vector for the delivery of GPC1 and assess its potential as an oral vaccination platform for cancer immunotherapy. Materials and Methods: EcN was genetically modified to produce a GPC1-flagellin fusion protein (GPC1-FL) to augment antigen immunogenicity. The expression and stability of GPC1 were confirmed in modified PANC02 cells using Western blot and flow cytometry, indicating that GPC1 expression did not influence tumor cell growth. A mouse model was employed to test immunogenicity post-oral delivery, measuring systemic IgG, IL-10, IL-2, and IFN-γ levels to indicate immune activation. Results: Oral immunization with EcN GPC1-FL elicited a robust systemic immune response, demonstrated by markedly increased levels of IgG and IL-10. IL-2 and IFN-γ concentrations were elevated in vaccinated mice relative to controls; however, the differences lacked statistical significance. Western blot examination of fecal samples verified consistent antigen expression in the gastrointestinal tract, indicating effective bacterial colonization and antigen retention. No detrimental impacts were noted, hence substantiating the safety of this methodology. Conclusions: These findings confirm EcN as a feasible and patient-friendly oral vaccination platform for cancer immunotherapy. The effective production of GPC1 in tumor cells, along with continuous antigen delivery and immune activation, underscores the promise of this approach for PDAC and other cancers. This study promotes microbial-based antigen delivery as a scalable, non-invasive substitute for traditional vaccine platforms.
Collapse
Affiliation(s)
- Idris Vruzhaj
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Marta Gambirasi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Aurora Giacomin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Amin Safa
- Doctoral School in Pharmacological Sciences, University of Padua, 35122 Padova, Italy
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
| |
Collapse
|
29
|
Morris DR, Qu Y, Haas de Mello A, Jones-Hall YL, Liu T, Weglarz M, Ivanciuc T, Garofalo RP, Casola A. Role of Hypoxia-Inducible Factors in Respiratory Syncytial Virus Infection-Associated Lung Disease. Int J Mol Sci 2025; 26:3182. [PMID: 40244000 PMCID: PMC11989216 DOI: 10.3390/ijms26073182] [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: 01/29/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are transcription factors that enable cells to adapt to low-oxygen environments. Viruses can exploit this pathway to enhance infection, making HIF modulation a potential antiviral strategy. In previous in vitro studies, we found that respiratory syncytial virus (RSV) stabilizes HIFs under normoxic conditions with inhibition of HIF-1α reducing replication. Despite several HIF-modulating compounds being tested or approved in other non-infectious models, little is known about their efficacy against respiratory viruses in relevant animal models. This study aimed to characterize the disease-modulating properties and antiviral potential of HIF-1α (PX478) and HIF-2α PT2385 inhibitors in RSV-infected BALB/c mice. We found that the inhibition of HIF-1α worsened clinical disease parameters while simultaneously improving airway function. Blocking HIF-1α also significantly reduced peak RSV replication in the lung. In contrast, the inhibition of HIF-2α was associated with improved clinical parameters, no changes in airway function, and reduced viral replication following RSV infection. The analysis of lung cells found significant modification in the T-cell compartment that correlated with changes in lung pathology and viral titers for each HIF inhibitor. This study underscores the differential roles of HIF proteins in RSV infection and highlights the need for further characterization of compounds currently in use or under therapeutic consideration.
Collapse
Affiliation(s)
- Dorothea R. Morris
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (D.R.M.); (M.W.); (R.P.G.)
- School of Population & Public Health, The University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.Q.); (A.H.d.M.); (T.L.); (T.I.)
| | - Yue Qu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.Q.); (A.H.d.M.); (T.L.); (T.I.)
| | - Aline Haas de Mello
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.Q.); (A.H.d.M.); (T.L.); (T.I.)
| | - Yava L. Jones-Hall
- School of Veterinary Medicine & Biomedical Science, Texas A&M University, College Station, TX 77843, USA;
| | - Tianshuang Liu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.Q.); (A.H.d.M.); (T.L.); (T.I.)
| | - Meredith Weglarz
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (D.R.M.); (M.W.); (R.P.G.)
| | - Teodora Ivanciuc
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.Q.); (A.H.d.M.); (T.L.); (T.I.)
| | - Roberto P. Garofalo
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (D.R.M.); (M.W.); (R.P.G.)
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.Q.); (A.H.d.M.); (T.L.); (T.I.)
| | - Antonella Casola
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (D.R.M.); (M.W.); (R.P.G.)
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.Q.); (A.H.d.M.); (T.L.); (T.I.)
| |
Collapse
|
30
|
Shanmugam G, George M, Sriram DK, Sarkar K. Exploring the regulatory role of mtTFA on inflammation, oxidative stress, and epigenetic alterations in COPD progressed NSCLC patients with smoking history. Int Immunopharmacol 2025; 150:114041. [PMID: 39965386 DOI: 10.1016/j.intimp.2025.114041] [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: 07/02/2024] [Revised: 11/17/2024] [Accepted: 01/05/2025] [Indexed: 02/20/2025]
Abstract
Oxidative stress and inflammation are pivotal in linking COPD to NSCLC progression, with dysregulated cytokine levels in inflamed COPD airways correlating with disease severity and lung cancer advancement. CD4+ T helper cells, pivotal in the inflammatory response, exhibit altered gene expression patterns in COPD and NSCLC, emphasizing their role in disease pathogenesis. Mitochondrial transcription factor A (mtTFA) dysregulation is implicated in inflammation and oxidative stress, affecting COPD and NSCLC progression. CRISPR/Cas9-mediated mtTFA depletion heightens inflammatory transcription factor expression, whileitsoverexpression reduces inflammation and oxidative stress markers. Histone deacetylases (HDACs) exhibit elevated expression in COPD and NSCLC CD4+ T cells, with inhibition via TSA treatment showing decreased inflammation and oxidative stress. Patients progressing from COPD to NSCLC demonstrate distinct epigeneticsignatures,with altered acetylation and methylation markers. TSA treatment enhances methylation and acetylation at the VEGFA gene locus, mitigating disease progression effects. Overexpression of mtTFA downregulates HDACs, while knockout upregulates them, influencing oxidative stress levels. These findings underscore the interplay between transcription factors, cytokines, and epigenetic regulation in CD4+ T cells, providing insights into disease mechanisms and potential therapeutic avenues. Understanding these pathways and targets may lead to innovative epigenetic therapies for COPD and NSCLC, potentially preventing COPD progression to NSCLC.
Collapse
Affiliation(s)
- Geetha Shanmugam
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Melvin George
- Department of Clinical Pharmacology, SRM Medical College Hospital and Research Centre, Kattankulathur, Tamil Nadu 603203, India
| | - Damal Kandadai Sriram
- Department of Endocrinology & Diabetology, Hindu Mission Hospital, West Tambaram, Chennai, Tamil Nadu 600045, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
31
|
Pan Y, Xue Y, Fei X, Zhao L, Han L, Su H, Lin Y, Zhou Y, Zhang Y, Xie G, Kong D, Bao W, Zhang M. PLK1 Mediates the Proliferation and Contraction of Airway Smooth Muscle Cells and Has a Role in T2-High Asthma with Neutrophilic Inflammation Model. J Inflamm Res 2025; 18:4381-4394. [PMID: 40162075 PMCID: PMC11954474 DOI: 10.2147/jir.s501645] [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: 12/17/2024] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
Background Type 2 (T2)-high asthma with neutrophilic inflammation is characterized by airway eosinophilic and neutrophilic infiltration, hyperresponsiveness, remodeling, and insensitivity to steroid treatment. Sphingosine-1-phosphate (S1P), which has a crucial role in the development of asthma, promotes the proliferation and contraction of airway smooth muscle cells (ASMCs), contributing to the pathophysiological processes of asthma. However, the downstream mediator of S1P remains unclear, as does its role in T2-high asthma with neutrophilic inflammation. Methods Ovalbumin- and ozone-induced murine models were used to replicate T2-high asthma with neutrophilic inflammation and primary ASMCs were applied to explore the underlying effects. Through transcriptomic analysis, PLK1 was identified as a potential key molecule associated with S1P-induced proliferation and contraction. Functional studies were performed both in vitro and in vivo by pharmacological inhibition to validate the role of PLK1 and to evaluate the therapeutic effects of PLK1 inhibition. Results S1P level was elevated in the bronchoalveolar lavage fluid (BALF) of T2-high asthma with neutrophilic inflammation model, and promoted ASMCs proliferation and contraction. PLK1 expression increased in S1P-stimulated ASMCs and asthmatic lung tissues. Inhibition of PLK1 blocked S1P-induced ASMCs proliferation and contraction. In vivo, PLK1 inhibition reduced airway inflammation (particularly neutrophilic infiltration), airway remodeling (airway smooth muscle proliferation and collagen deposition), and airway hyperresponsiveness and resistance, improving lung function (of both large and small airways), with superior therapeutic effects to those of dexamethasone. In addition, PLK1 inhibition markedly reduced the BALF levels of IL-17A, IL-21 and IL-6, suggesting that PLK1 might exert its effects mainly through the regulation of Th17 pathway. Conclusion PLK1 mediates S1P-induced ASMC proliferation and contraction, and plays an important part in T2-high asthma with neutrophilic inflammation model, making it a potential therapeutic target for treating T2-high asthma with neutrophilic inflammation.
Collapse
Affiliation(s)
- Yilin Pan
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yishu Xue
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xia Fei
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Lei Zhao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Lei Han
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Hang Su
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yanmei Lin
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yan Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yingying Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Guogang Xie
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Deping Kong
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Wuping Bao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
32
|
Zhou Q, Jing M, Ren H, Li G, Wang Z. Efficacy of electroacupuncture on clinical signs and immunological factors in herpes zoster: The first systematic review, meta-analysis, and trial sequential analysis of randomized clinical trials. Medicine (Baltimore) 2025; 104:e41458. [PMID: 40128056 PMCID: PMC11936656 DOI: 10.1097/md.0000000000041458] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 01/17/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Electroacupuncture (EA) is utilized to address various health conditions. Herein, we designed a systematic review and meta-analysis to evaluate the efficacy of EA on clinical and immunological factors in herpes zoster (HZ) based on randomized clinical trials. METHODS Four international databases and 3 Chinese databases were searched until January 2024. We used RevMan 5.3 for meta-analysis and presented the data as standardized mean difference (SMD) or odds ratio (OR) and 95% confidence interval. RESULTS A total of 1361 records were identified in the databases and at last, 19 articles were entered into the meta-analysis. The result shows a negative pooled SMD of -2.55 (P < .00001) for the VAS score. The pooled SMD for cessation of pustules time in the case group compared to the control group was -0.69 (P = .0008), for pain relief time was -1.36 (P = .002), for the time to scab was -0.47 (P = .009), and for time to remove scab was -1.01 (P = .0003). The pooled OR for the incidence of postherpetic neuralgia was 0.11 (P < .00001), and the total effective rate was 4.25 (P < .00001). The pooled SMD for the cluster of differentiation (CD)3 count was 2.59 (P = .07), for the CD4 count was 2.81 (P = .04), for the CD8 count was -0.75 (P = .50), and for theCD4/CD8 ratio was 1.12 (P = .15). CONCLUSIONS The results indicate that the EA treatment had several significant benefits compared to Western medicine (WM) in HZ patients in terms of clinical and immunological factors. But, the combination of treatments of EA with WM had better effects compared to EA treatment alone.
Collapse
Affiliation(s)
- Qiaoli Zhou
- Department of Nursing, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mao Jing
- Department of Acupuncture and moxibustion and Trauma College of Hubei University of Traditional Chinese Medicine, Wuhan City, Hubei Province, China
| | - Haitao Ren
- School of Life and Health of Huzhou College, Huzhou City, Zhejiang Province, China
| | - Gaokai Li
- School of Life and Health of Huzhou College, Huzhou City, Zhejiang Province, China
| | - Zongjiao Wang
- Fitness Teaching and Research Office, Professional Tennis Academy, Wuhan Urban Vocational College, Wuhan City, Hubei Province, China
| |
Collapse
|
33
|
Chegene Lorestani R, A Ahmad T, Heidarinia H, Goudarzi F, Khaledian S, Ghadiri K, Rostamian M. Computational design of a fimbriae-derived multi-epitope vaccine candidate against Klebsiella pneumoniae. J Biomol Struct Dyn 2025:1-17. [PMID: 40056379 DOI: 10.1080/07391102.2025.2472407] [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: 11/06/2023] [Accepted: 04/10/2024] [Indexed: 03/10/2025]
Abstract
Klebsiella pneumoniae is a pathogen that causes infections in various parts of the body, with high mortality rates reported in antibiotic-resistant cases. Treating at-risk individuals requires crucial vaccination efforts due to the challenges that exist. This research involved designing a multi-epitope vaccine from K. pneumoniae's fimbriae antigens. Optimal T-cell and B-cell epitopes were chosen through in silico studies including epitope-HLAs molecular docking. The multi-epitope was created, featuring antigenic T- and B-cell epitopes, β-defensin as an adjuvant, the PADRE sequence to boost immunogenicity and well-suited linkers. The tertiary structure of the multi-epitope was achieved through modeling and molecular dynamics-based refinements. The construct underwent scrutiny for structural traits, physicochemical properties, conformational B epitope prediction, immune responses simulation, in silico cloning, molecular docking for assay binding to toll-like receptors (TLRs), and deformability studies. The outcomes indicated the vaccine candidate's positive attributes, encompassing immunogenicity, structure, physicochemical properties, solubility, TLR binding, toxicity, stability, allergenicity, and cross-reactivity. The multi-epitope vaccine candidate exhibits the potential for provoking diverse immune responses against K. pneumoniae. Nevertheless, additional in vitro and in vivo experimental tests are necessary to substantiate its efficacy.
Collapse
Affiliation(s)
- Roya Chegene Lorestani
- Infectious Diseases Research Center, Health Policy and Promotion Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Tarek A Ahmad
- Library Sector, Bibliotheca Alexandrina, Alexandria, Egypt
| | - Hana Heidarinia
- Department of Microbiology, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Farjam Goudarzi
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Salar Khaledian
- Infectious Diseases Research Center, Health Policy and Promotion Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Keyghobad Ghadiri
- Infectious Diseases Research Center, Health Policy and Promotion Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mosayeb Rostamian
- Infectious Diseases Research Center, Health Policy and Promotion Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
34
|
Tschongov T, Konwar S, Kleindienst J, Dabrowska-Schlepp P, Busch A, Schaaf A, Schell C, Rogg M, Häffner K. Effective long-term treatment with moss-produced factor H by overcoming the antibody response in a mouse model of C3G. Front Immunol 2025; 16:1535547. [PMID: 40124383 PMCID: PMC11925764 DOI: 10.3389/fimmu.2025.1535547] [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: 11/27/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Complement-associated disorders are caused by the dysregulation and disbalance of the complement system, especially excessive activation. Most drugs that target the complement system are designed to inhibit the complement pathway at either the proximal or terminal levels. The use of a natural complement regulator such as factor H (FH) could provide a superior treatment option by restoring balance to an overactive complement system. We recently reported the moss-based production of an analog of human FH with an optimized glycan profile (CPV-104), which showed in vitro and in vivo characteristics comparable to its human counterpart. Here, we follow up our previous work, focusing in more detail on the time course and long-term efficacy of CPV-104 treatment in FH-deficient (FH -/-) mice. The analysis of long-term treatment effects following multiple injections of human FH into mice was previously hindered by the immune response, so we developed a protocol for the sustained depletion of CD20+ B-cells and CD4+ T-cells, preventing antibody formation without influencing the C3G phenotype. Using this dual-depletion method, we were able to complete dosing interval experiments in FH -/- mice, administering up to three injections of CPV-104 at different intervals. Repeated CPV-104 administration was able to lastingly resolve C3 deposits, offering additional rationale for the clinical testing of CPV-104 in human C3G patients. Moreover, our novel dual-depletion method has the potential for adaptation to different mouse models, allowing the testing of multiple doses of other therapeutic proteins.
Collapse
Affiliation(s)
- Todor Tschongov
- Department of Internal Medicine IV, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Swagata Konwar
- Department of Internal Medicine IV, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Biology, Albert-Ludwig University Freiburg, Freiburg, Germany
| | - Jessika Kleindienst
- Department of Internal Medicine IV, Medical Faculty, University of Freiburg, Freiburg, Germany
| | | | - Andreas Busch
- Nonclinical Development, Eleva GmbH, Freiburg, Germany
| | | | - Christoph Schell
- Institute for Surgical Pathology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Manuel Rogg
- Institute for Surgical Pathology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Karsten Häffner
- Department of Internal Medicine IV, Medical Faculty, University of Freiburg, Freiburg, Germany
| |
Collapse
|
35
|
Yuan VG, Xia A, Santa Maria PL. Chronic suppurative otitis media: disrupted host-microbial interactions and immune dysregulation. Front Immunol 2025; 16:1547206. [PMID: 40114926 PMCID: PMC11923626 DOI: 10.3389/fimmu.2025.1547206] [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: 12/17/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Recent research has uncovered new mechanisms that disrupt the balance between the host and microbes in the middle ear, potentially leading to dysbiosis and chronic suppurative otitis media (CSOM). Dysbiotic microbial communities, including core pathogens such as persister cells, are recognized for displaying cooperative virulence. These microbial communities not only evade the host's immune defenses but also promote inflammation that leads to tissue damage. This leads to uncontrolled disorder and pathogen proliferation, potentially causing hearing loss and systemic complications. In this discussion, we examine emerging paradigms in the study of CSOM that could provide insights into other polymicrobial inflammatory diseases. Additionally, we underscore critical knowledge gaps essential for developing a comprehensive understanding of how microbes interact with both the innate and adaptive immune systems to trigger and maintain CSOM.
Collapse
Affiliation(s)
- Vincent G. Yuan
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, United States
| | - Anping Xia
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, United States
| | - Peter L. Santa Maria
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, United States
| |
Collapse
|
36
|
Pang Y, Wang C, Zhang YZ, Wang Z, Imoto S, Lee TY. STForte: tissue context-specific encoding and consistency-aware spatial imputation for spatially resolved transcriptomics. Brief Bioinform 2025; 26:bbaf174. [PMID: 40254832 PMCID: PMC12009714 DOI: 10.1093/bib/bbaf174] [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: 01/08/2025] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Encoding spatially resolved transcriptomics (SRT) data serves to identify the biological semantics of RNA expression within the tissue while preserving spatial characteristics. Depending on the analytical scenario, one may focus on different contextual structures of tissues. For instance, anatomical regions reveal consistent patterns by focusing on spatial homogeneity, while elucidating complex tumor micro-environments requires more expression heterogeneity. However, current spatial encoding methods lack consideration of the tissue context. Meanwhile, most developed SRT technologies are still limited in providing exact patterns of intact tissues due to limitations such as low resolution or missed measurements. Here, we propose STForte, a novel pairwise graph autoencoder-based approach with cross-reconstruction and adversarial distribution matching, to model the spatial homogeneity and expression heterogeneity of SRT data. STForte extracts interpretable latent encodings, enabling downstream analysis by accurately portraying various tissue contexts. Moreover, STForte allows spatial imputation using only spatial consistency to restore the biological patterns of unobserved locations or low-quality cells, thereby providing fine-grained views to enhance the SRT analysis. Extensive evaluations of datasets under different scenarios and SRT platforms demonstrate that STForte is a scalable and versatile tool for providing enhanced insights into spatial data analysis.
Collapse
Affiliation(s)
- Yuxuan Pang
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Chunxuan Wang
- School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
| | - Yao-zhong Zhang
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Zhuo Wang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, No. 75 Bo-Ai Street, Hsinchu 300, Taiwan
| |
Collapse
|
37
|
Kline EM, Jernigan JE, Scharer CD, Maurer J, Hicks SL S, Herrick M MK, Wallings RL, Kelly SD, Chang J, Menees KB, McFarland NR, Boss JM, Tansey MG, Joers V. MHCII reduction is insufficient to protect mice from alpha-synuclein-induced degeneration and the Parkinson's HLA locus exhibits epigenetic regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.31.610581. [PMID: 40093159 PMCID: PMC11908218 DOI: 10.1101/2024.08.31.610581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Major histocompatibility complex class II (MHCII) molecules are antigen presentation proteins and increased in post-mortem Parkinson's disease (PD) brain. Attempts to decrease MHCII expression have led to neuroprotection in PD mouse models. Our group reported that a SNP at rs3129882 in the MHCII gene Human leukocyte Antigen (HLA) DRA is associated with increased MHCII transcripts and surface protein and increased risk for late-onset idiopathic PD. We therefore hypothesized that decreased MHCII may mitigate dopaminergic degeneration. During an ongoing α-synuclein lesion, mice with MHCII reduction in systemic and brain innate immune cells (LysMCre+I-Abfl/fl or CRE+) displayed brain T cell repertoire shifts and greater preservation of the dopaminergic phenotype in nigrostriatal terminals. Next, we investigated a human cohort to characterize the immunophenotype of subjects with and without the high-risk GG genotype at the rs3129882 SNP. We confirmed that the high-risk GG genotype is associated with peripheral changes in MHCII inducibility, frequency of CD4+ T cells, and differentially accessible chromatin regions within the MHCII locus. Although our mouse studies indicate that myeloid MHCII reduction coinciding with an intact adaptive immune system is insufficient to fully protect dopamine neurons from α-synuclein-induced degeneration, our data are consistent with the overwhelming evidence implicating antigen presentation in PD pathophysiology.
Collapse
Affiliation(s)
- Elizabeth M Kline
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT USA
| | - Janna E Jernigan
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA USA
| | - Jeffrey Maurer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA USA
| | - Sakeenah Hicks SL
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA USA
| | - Mary K Herrick M
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL USA
| | - Rebecca L Wallings
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL USA
| | - Sean D Kelly
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jianjun Chang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelly B Menees
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL USA
| | - Nikolaus R McFarland
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL USA
- Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA USA
| | - Malú Gámez Tansey
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL USA
- Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL USA
- McKnight Brain Institute, University of Florida Health, Gainesville, FL USA
| | - Valerie Joers
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL USA
- McKnight Brain Institute, University of Florida Health, Gainesville, FL USA
| |
Collapse
|
38
|
Patel P, Patel B, Vyas SD, Patel MS, Hirani T, Haque M, Kumar S. A Narrative Review of Periodontal Vaccines: Hope or Hype? Cureus 2025; 17:e80636. [PMID: 40091902 PMCID: PMC11910667 DOI: 10.7759/cureus.80636] [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: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 03/19/2025] Open
Abstract
Globally, periodontal diseases, mainly driven by polymicrobial biofilms, are a widespread concern of social medicine due to their considerable incidence and tie-up to systemic disorders like diabetes, cardiovascular diseases, and complications during pregnancy. Traditional treatments focus on mechanical debridement and antimicrobial therapies, but these approaches have limitations, including recurrence and antibiotic resistance. Periodontal vaccines offer a promising alternative by targeting the immunological mechanisms underlying periodontal disease. This review explores the current state of periodontal vaccine development, highlighting key antigens, vaccine delivery systems, and preclinical and clinical advancements. Special emphasis is placed on antigen selection, host variability, immune tolerance, and future directions to overcome these barriers. This article highlights the advancements and challenges in periodontal vaccine research, offering insights into the capability of immunoprophylaxis as a groundbreaking way to manage periodontal diseases.
Collapse
Affiliation(s)
- Pratiksha Patel
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Bhavin Patel
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Shruti D Vyas
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Maitri S Patel
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Tanvi Hirani
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Mainul Haque
- Department of Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
- Department of Research, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Santosh Kumar
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| |
Collapse
|
39
|
Alrashdi BM, Askar H, Germoush MO, Fouda M, Massoud D, Alzwain S, Abdelsater N, Salim LMS, Gadelmawla MHA, Ashry M. Cardioprotective, anti-inflammatory, and antioxidative outcome of costus against bleomycin-induced cardiotoxicity in rat model. J Genet Eng Biotechnol 2025; 23:100466. [PMID: 40074440 PMCID: PMC11904485 DOI: 10.1016/j.jgeb.2025.100466] [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: 10/17/2024] [Revised: 12/27/2024] [Accepted: 02/01/2025] [Indexed: 03/14/2025]
Abstract
Due to bleomycin's cytotoxic characteristics, which include cardiotoxicity, this investigation looked at the effectiveness of costus ethanolic extract in reducing cardiotoxicity in male rats receiving bleomycin therapy. Forty adult male rats (160-200 g) were evenly allocated into four groups: group (1) included normal rats serving as the control; group (2) included normal rats administered 200 mg/kg of costus ethanolic extract (CEE) orally for 6 weeks; group (3) consisted of rats receiving bleomycin (15 mg/kg twice weekly, ip) for 6 weeks; and group (4) involved rats treated orally with CEE (200 mg/kg/day) for 6 weeks following bleomycin intoxication. The results indicated that the CEE significantly reversed the cardiological deteriorations brought on by bleomycin; this was demonstrated by a considerable increase in cardiac SOD, GPx, GSH, and CAT, along with a substantial decrease in cardiac MDA, NO, and DNA fragmentation. Also, serum, LDH, CK-MB, CK- total, TNF-α, IL-4, IL-6 IL-10, IL-1β, triglycerides, cholesterol, and LDL were significantly reduced, while CD4 levels increased, and HDL declined significantly. The results of the histological and immunohistochemical analyses revealed a notable regeneration. In conclusion, CEE's anti-cardiotoxic, anti-inflammatory, and antioxidant properties prove its ability to be a cardio-protective supplement. This may be mediated by its active constituents' radical scavenging and antioxidant properties, particularly high phenolic content.
Collapse
Affiliation(s)
- Barakat M Alrashdi
- Biology Department, College of Science, Jouf University, P.O. Box: 2014, Sakaka, Saudi Arabia.
| | - Hussam Askar
- Zoology Department, Faculty of Science, Al-Azhar University, 71524 Assiut, Egypt.
| | - Mousa O Germoush
- Biology Department, College of Science, Jouf University, P.O. Box: 2014, Sakaka, Saudi Arabia
| | - Maged Fouda
- Biology Department, College of Science, Jouf University, P.O. Box: 2014, Sakaka, Saudi Arabia
| | - Diaa Massoud
- Biology Department, College of Science, Jouf University, P.O. Box: 2014, Sakaka, Saudi Arabia
| | - Sarah Alzwain
- Biology Department, College of Science, Jouf University, P.O. Box: 2014, Sakaka, Saudi Arabia
| | - Naser Abdelsater
- Zoology Department, Faculty of Science, Al-Azhar University, 71524 Assiut, Egypt
| | - Laila M S Salim
- Nutration and Food Science, Faculty of Specific Education, Assiut University, Assiut, Egypt
| | | | - Mahmoud Ashry
- Zoology Department, Faculty of Science, Al-Azhar University, 71524 Assiut, Egypt
| |
Collapse
|
40
|
Vuscan P, Röring RJ, Kischkel B, Tintoré M, Cuñé J, de Lecea C, Joosten LAB, Netea MG. Effect of Saccharomyces cerevisiae β-glucan on the T helper cytokine profile. Cytokine 2025; 187:156871. [PMID: 39889564 DOI: 10.1016/j.cyto.2025.156871] [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: 07/08/2024] [Revised: 11/27/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025]
Abstract
ABBi16 is a high-complexity blend of β-1,3/1,6-glucans from Saccharomyces cerevisiae with strong immunomodulatory activities, that have been recently shown to support anti-tumoral immune responses through the induction of trained immunity. Whether ABBi16 also modulates the balance between the various T helper (Th) lymphocyte responses is not known. Here, we show that ABBi16 induces Th1 responses, as indicated by stimulation of IFNγ and TNF production by human peripheral blood mononuclear cells (PBMCs). Moreover, the elevated secretion of IL-10 and IL-22 suggests a potential regulatory response of the Th1/Th2/Th17 balance. Co-stimulating PBMCs with ABBi16 alongside Bacille Calmette-Guerin (BCG), IL-1beta + IL-23, and IL-12 + IL-18 cytokine combinations further enhanced Th1 polarization and IL-22 induction, hinting at an additive effect of β-glucan on both Th1 and regulatory Th17 immune responses. ABBi16 did not induce IL-17 production, the prototype pro-inflammatory product of Th17 responses, suggesting that it can be safely used as an oral supplement in patients with autoimmune conditions. These results highlight the potential of ABBi16 to regulate the Th1/Th2/Th17 balance toward antimicrobial and regulatory effects.
Collapse
Affiliation(s)
- Patricia Vuscan
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rutger J Röring
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Brenda Kischkel
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Maria Tintoré
- AB Biotek Human Nutrition and Health, Barcelona, Spain
| | - Jordi Cuñé
- AB Biotek Human Nutrition and Health, Barcelona, Spain
| | | | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany.
| |
Collapse
|
41
|
Tirelli E, Pucci M, Squillario M, Bignotti G, Messali S, Zini S, Bugatti M, Cadei M, Memo M, Caruso A, Fiorentini S, Villanacci V, Uberti D, Abate G. Effects of methylglyoxal on intestine and microbiome composition in aged mice. Food Chem Toxicol 2025; 197:115276. [PMID: 39863075 DOI: 10.1016/j.fct.2025.115276] [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: 08/06/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Methylglyoxal (MGO), a highly reactive precursor of advanced glycation end products (AGEs), is endogenously produced and prevalent in various ultra-processed foods. MGO has emerged as a significant precursor implicated in the pathogenesis of type 2 diabetes and neurodegenerative diseases. To date, the effects of dietary MGO on the intestine have been limited explored. Thus, this study investigates the impact of prolonged oral administration of MGOs on gut health in aged mice. METHODS Aged mice received MGO chronically (100 mg/kg/day) for 4 weeks Intestinal samples were analyzed using RT-PCR and immunohistochemistry for proinflammatory cytokines, permeability markers, and tight junction proteins. 16S rRNA gene-based microbiome analysis was also performed to characterize microbiome composition and its metabolic potential. RESULTS MGO treatment induced notable alterations at the intestinal level, characterized by an increased formation of MGO-glycated proteins with a concurrent induction of a pro-inflammatory status and reduced expression and delocalization of zonulin-1 and occludin, tight junction proteins. Changes in intestinal morphology were also observed, including hyperproliferation of Paneth cells and an augmented thickness of the intestinal mucus layer, as indicated by immunohistochemical data from MGO-treated mice. Investigation into the microbiota composition revealed that MGO is effective in selectively modifying its composition and metabolic pathways. A decreased abundance of bacterial genera associated with the production of acetic and butyric acids (i.e. Harryflintia, Intestinimonas and Ruminococcaceae genera) and a substantial increase in Lachnospiraceae and Akkermansia genera were found in MGO-treated mice. CONCLUSION These findings highlight how dietary MGO can affect intestinal balance, providing valuable insights into the potential links between glycotoxins, gut microbiota, and overall gut functionality.
Collapse
Affiliation(s)
- Emanuela Tirelli
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Mariachiara Pucci
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | | | - Gloria Bignotti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Serena Messali
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Stefania Zini
- Institute of Pathology, Spedali Civili di Brescia, Italy
| | - Mattia Bugatti
- Institute of Pathology, Spedali Civili di Brescia, Italy
| | - Moris Cadei
- Institute of Pathology, Spedali Civili di Brescia, Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Simona Fiorentini
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Italy
| | | | - Daniela Uberti
- Department of Molecular and Translational Medicine, University of Brescia, Italy.
| | - Giulia Abate
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| |
Collapse
|
42
|
Kaur K, Jewett A. Distinct profiles of osteoclast and dendritic cell-mediated expansion and functional activation of NK and T cells. Cancer Immunol Immunother 2025; 74:127. [PMID: 40024920 PMCID: PMC11872835 DOI: 10.1007/s00262-025-03956-x] [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: 09/03/2024] [Accepted: 01/22/2025] [Indexed: 03/04/2025]
Abstract
Osteoclasts (OCs) and dendritic cells (DCs) induce expansion and functional activation of NK and T cells. When comparing OCs with DC-induced activation in NK cells, OCs induced significantly higher cell expansion and functional activation of NK cells as compared to DCs, either from healthy individuals or those obtained from cancer patients. However, no differences could be seen in the levels of cell expansion and functional activation in T cells activated by OCs or DCs, either from healthy individuals or those from cancer patients. OCs selectively expanded and activated CD8 + T cells, whereas DCs expanded and activated CD4 + T cells. In addition, both allogeneic and autologous OCs induced similar levels of cell expansion and functional activation of NK and T cells. Together, these findings highlighted the essential roles of OCs in expanding and activating the cytotoxic effectors of NK, and CD8 + T cells, and demonstrated several differences when compared to the effect of DCs.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA.
- The Jonsson Comprehensive Cancer Center, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA.
| |
Collapse
|
43
|
Gong Z, Xu H, Zhang Q, Wang G, Fan L, Wang Z, Fan L, Liu C, Yu Y, Liu Z, Zhou Q, Xiao H, Hou R, Zhao Y, Chen Y, Xie J. Unveiling the immunological landscape of disseminated tuberculosis: a single-cell transcriptome perspective. Front Immunol 2025; 16:1527592. [PMID: 40092995 PMCID: PMC11906432 DOI: 10.3389/fimmu.2025.1527592] [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: 11/13/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Hematogenous disseminated tuberculosis (DTB) has an unclear etiology that likely involves multiple factors. Understanding the underlying immunological characteristics of DTB is crucial for elucidating its pathogenesis. Methods We conducted single-cell RNA transcriptome and T cell receptor (TCR) sequencing on samples from seven DTB patients. Additionally, we integrated and analyzed data from two published profiles of latent TB infection, three active TB cases, and two healthy controls. Results Our analysis revealed a significantly higher proportion of inflammatory immune cells (e.g., monocytes and macrophages) in DTB patients, along with a notably lower abundance of various lymphocytes (including T cells, B cells, and plasma cells), suggesting that lymphopenia is a prominent feature of the disease. T cell pseudotime analysis indicated a decrease in the expression of most hypervariable genes over time, pointing to T cell functional exhaustion. Furthermore, a marked absence of mucosal-associated invariant T (MAIT) cells was observed in the peripheral blood of DTB patients. In the TCR repertoire, specific polymorphisms (TRAV9-2, TRAV13-1, TRBV20-1, and TRBV5-1) and dominant clones (TRAJ49, TRBJ2-7, and TRBJ2-1) were identified. Analysis of the complementarity determining region 3 (CDR3) showed that the most frequent combination was TRAV1-2/TRAJ33, with the motif "CAAMD" being significantly reduced in DTB patients. Discussion These findings suggest that lymphopenia and T cell exhaustion, along with unique TCR signatures, may play critical roles in DTB pathogenesis. The reduced "CAAMD" motif and altered TCR clonotypes provide novel insights into the complex cellular dynamics associated with the disease, potentially offering new avenues for targeted immunological interventions.
Collapse
Affiliation(s)
- Zhen Gong
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hongxiang Xu
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Qiao Zhang
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Guirong Wang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Lin Fan
- Shanghai Clinical Research Center for Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zilu Wang
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Lichao Fan
- Shenyang Tenth People’s Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, China
| | - Chang Liu
- Shenyang Tenth People’s Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, China
| | - Yanhong Yu
- Shenyang Tenth People’s Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, China
| | - Zhou Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qiang Zhou
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | | | - Rui Hou
- Shanghai Biotechnology Corporation, Shanghai, China
| | - Ying Zhao
- Shanghai Biotechnology Corporation, Shanghai, China
| | - Yu Chen
- Shenyang Tenth People’s Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| |
Collapse
|
44
|
Maciocia N, Wade B, Maciocia P. CAR T-cell therapies for T-cell malignancies: does cellular immunotherapy represent the best chance of cure? Blood Adv 2025; 9:913-923. [PMID: 39715467 PMCID: PMC11876835 DOI: 10.1182/bloodadvances.2023012263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/31/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024] Open
Abstract
ABSTRACT Chimeric antigen receptor T-cell (CAR-T) therapy has proven successful for B-cell lymphomas and leukemias. This success has inspired the development of CAR-T for T-cell malignancies. T-cell lymphomas and T-cell acute lymphoblastic leukemia (T-ALL) are highly heterogenous diseases but are united by poor prognosis in the relapsed/refractory setting and the lack of any novel, targeted therapies. CAR-T therapy is a promising solution for these diseases but carries a number of challenges, principally that target antigens are typically shared between malignant and normal T cells. This can cause issues with fratricide and T-cell aplasia. In this review we discuss the current state of CAR-T treatment for T-ALL and T-cell lymphomas, highlighting recent novel clinical data for T-cell malignancies and discuss lessons that can be learned for future research in this area.
Collapse
Affiliation(s)
- Nicola Maciocia
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Brandon Wade
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Paul Maciocia
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| |
Collapse
|
45
|
Yuk CM, Hong S, Kim D, Kim M, Jeong HW, Park SJ, Min H, Kim W, Lim J, Kim HD, Kim SG, Seong RH, Kim S, Lee SH. Inositol polyphosphate multikinase regulates Th1 and Th17 cell differentiation by controlling Akt-mTOR signaling. Cell Rep 2025; 44:115281. [PMID: 39946233 DOI: 10.1016/j.celrep.2025.115281] [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: 08/08/2023] [Revised: 11/15/2024] [Accepted: 01/16/2025] [Indexed: 02/28/2025] Open
Abstract
Activated proinflammatory T helper (Th) cells, including Th1 and Th17 cells, drive immune responses against pathogens and contribute to autoimmune diseases. We show that the expression of inositol polyphosphate multikinase (IPMK), an enzyme essential for inositol phosphate metabolism, is highly induced in Th1 and Th17 subsets. Deletion of IPMK in CD4+ T cells leads to diminished Th1- and Th17-mediated responses, reducing resistance to Leishmania major and attenuating experimental autoimmune encephalomyelitis. IPMK-deficient CD4+ T cells show impaired activation and Th17 differentiation, linked to the decreased activation of Akt, mTOR, and STAT3. Mechanistically, IPMK functions as a phosphatidylinositol 3-kinase to regulate phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) production, promoting T cell activation and effector functions. In IPMK-deficient CD4+ T cells, T cell receptor-stimulated PtdIns(3,4,5)P3 generation is abolished by wortmannin, suggesting IPMK acts in a wortmannin-sensitive manner. These findings establish IPMK as a critical regulator of Th1 and Th17 differentiation, underscoring its role in maintaining immune homeostasis.
Collapse
Affiliation(s)
- Chae Min Yuk
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Sehoon Hong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Dongeon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea; Biomedical Research Center, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; VA Palo Alto Health Care System, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mingyo Kim
- Division of Rheumatology, Department of Internal Medicine, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; Faculty of Medicine, University of Münster, 48149 Münster, Germany
| | - Seung Ju Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Hyungyu Min
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Republic of Korea
| | - Wooseob Kim
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Republic of Korea
| | - Jongbu Lim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Hyo Dam Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Sang-Gyu Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Rho Hyun Seong
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Republic of Korea.
| | - Seyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea; KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea.
| | - Seung-Hyo Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea; Biomedical Research Center, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-Gu, Daejeon 34141, Republic of Korea; Department of Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
46
|
Bu S, Liu M, Yang L, Lee P, Miller H, Park CS, Byazrova M, Filatov A, Benlagha K, Gaber T, Buttgereit F, Gong Q, Zhai Z, Liu C. The function of T cells in immune thrombocytopenia. Front Immunol 2025; 16:1499014. [PMID: 40061938 PMCID: PMC11885273 DOI: 10.3389/fimmu.2025.1499014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 05/13/2025] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease, characterized by increased bleeding due to a reduced platelet count. The pathogenesis of ITP is very complex and involves autoantibody production and T-cell-mediated immune abnormalities. An imbalance of effector and regulatory CD4+ T cells and the breach of tolerance primarily cause ITP, leading to the dysfunctional development of autoreactive Th cells (including Th1, Th2, and Th17 cells) and Tregs. The loss of auto-platelet antigen tolerance in ITP results in autoantibody- and cytotoxic T-cell-mediated platelet clearance. T-cell-related genetic risk factors significantly influence the development and progression of this disease. New therapies targeting T cells have emerged as potentially effective cures for this disease. This review summarizes the role of T cells in ITP.
Collapse
Affiliation(s)
- Siyuan Bu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Liu
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Lu Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Chan-Sik Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Maria Byazrova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Alexander Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Kamel Benlagha
- Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
47
|
Kuo CY, Huang CY, Chen HM, Chen LC, Kuo ML. Antagonism of CD28 blocks allergic responses in the ovalbumin-induced asthmatic model mice. Int Immunopharmacol 2025; 148:114071. [PMID: 39842142 DOI: 10.1016/j.intimp.2025.114071] [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: 06/04/2024] [Revised: 10/30/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025]
Abstract
Allergen-reactive T helper (Th) 2 cells play a pivotal role in initiating asthma pathogenesis. The absence or interruption of CD28 signaling causes significant consequences for T-cell activation, leading to reduced cell proliferation and interleukin (IL)-2 production. A novel compound, Cyn-1324, exhibits a higher binding affinity to CD28 than CD80. Thus, targeting the CD28-CD80 interaction emerges as a promising therapeutic approach for allergic asthma. However, the impact of CD28 antagonists on allergen-induced asthma remains unreported. In this study, we explored the effects of intranasally administered Cyn-1324 on airway inflammation in the ovalbumin (OVA)-induced murine allergic model. The results revealed a significant reduction in airway hyper-responsiveness (AHR), eosinophil recruitment, and cell infiltration in lung tissues, as well as decreased OVA-specific IgE in serum and Th2 cytokine levels in OVA-stimulated lymphocyte cultures. Additionally, we demonstrated the immunosuppressive effects of Cyn-1324 in vitro, including decreased T-cell proliferation and IL-2 secretion, together with increased p27kip1 expression via inhibiting the PI3K signaling pathway. Notably, Cyn-1324 not only inhibited the NF-κB pathway, but also appeared to suppress p38 activation, which is downstream of CD3 signaling, and reduced calcium-induced NFAT protein expression. These findings suggest that Cyn-1324 alleviates allergic responses by inhibiting the CD28-CD80 interaction and holds promise as an immunosuppressive agent for allergic patients.
Collapse
Affiliation(s)
- Chieh-Ying Kuo
- Department of Microbiology and Immunology Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan
| | - Chih-Yu Huang
- Department of Microbiology and Immunology Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan
| | - Hueih-Min Chen
- Taiwan Semiconductor Research Institute National Applied Research Laboratories Hsinchu Taiwan
| | - Li-Chen Chen
- Division of Allergy Asthma and Rheumatology Department of Pediatrics Chang Gung Memorial Hospital at Linkou Taoyuan Taiwan; Department of Pediatrics New Taipei Municipal TuCheng Hospital New Taipei Taiwan
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan; Division of Allergy Asthma and Rheumatology Department of Pediatrics Chang Gung Memorial Hospital at Linkou Taoyuan Taiwan; Department of Pediatrics New Taipei Municipal TuCheng Hospital New Taipei Taiwan; Research Center for Chinese Herbal Medicine College of Human Ecology Chang Gung University of Science and Technology Taoyuan Taiwan.
| |
Collapse
|
48
|
Jia J, Guo J, Yan C, Gu Y, Xia X. Oyster powder supplementation enhances immune function in mice partly through modulating the gut microbiota and arginine metabolism. Food Funct 2025; 16:1254-1266. [PMID: 39868593 DOI: 10.1039/d4fo06068g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Oysters are well-known for their health benefits such as immuno-modulatory functions. The intestinal microbiome serves as a key mediator between diet and immune regulation. This study aimed to investigate whether oyster consumption could alleviate cyclophosphamide (Cy)-induced immunosuppression by promoting intestinal homeostasis. In mice treated with Cy, a significant decrease in immune cells and cytokines was observed. In contrast, mice supplemented with oyster powder demonstrated elevated numbers of immune cells in the spleen and small intestine, as well as enhanced serum production of IL-1β, IL-2, TNF-α, and IFN-γ. Furthermore, oyster consumption improved the composition of the gut microbiota by promoting beneficial bacteria and inhibiting harmful ones. Metabolomics analysis revealed that oyster powder treatment significantly enhanced the arginine biosynthesis pathway, and further analysis found that the consumption of oysters led to increased arginine levels. Correlation analysis showed a significant positive correlation between L-arginine and immune-related markers. Collectively, these findings suggest that oyster consumption may enhance immunity by modulating the gut microbiota and boosting arginine biosynthesis pathways. Dietary oyster consumption could be an effective strategy to support immune health.
Collapse
Affiliation(s)
- Jinhui Jia
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| | - Jian Guo
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| | - Chunhong Yan
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| | - Yunqi Gu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| | - Xiaodong Xia
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| |
Collapse
|
49
|
Silva DKC, Novo LBDC, Ribeiro IM, Barreto BC, Opretzka LCF, Meira CS, Soares MBP. Physalin F, a Potent Inhibitor of Lymphocyte Function, Is a Calcineurin Inhibitor and Has Synergistic Effect with Dexamethasone. Molecules 2025; 30:916. [PMID: 40005226 PMCID: PMC11858416 DOI: 10.3390/molecules30040916] [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: 01/04/2025] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
The dysregulation of immune responses are responsible for the development of several diseases, such as allergic and autoimmune diseases. The medications used to treat these conditions have numerous side effects, creating the need for new drugs. Physalins are natural compounds with various pharmacological activities already described. Here, we aimed to investigate the immunomodulatory effects of physalin F in mouse splenocytes and in a delayed-type hypersensitivity (DTH) model. In a cytotoxicity assay, physalin F had low cytotoxicity to mouse splenocytes in concentrations equal to or below 2 µM. It significantly inhibited lymphocyte proliferation in a concentration-dependent manner and reduced the production of cytokines, including IL-2, IL-4, IL-10, and IFN-γ, in activated splenocytes. The combined therapy of physalin F with dexamethasone was investigated in vitro, showing a synergistic action of the two compounds. Mechanistically, physalin F reduced calcineurin activity in concanavalin A-stimulated splenocyte cultures. Finally, in vivo, the intraperitoneal administration of physalin F in a DTH model reduced paw edema induced by bovine serum albumin immunization. Our results demonstrate the potential of physalin F as an immunosuppressive agent, to be used alone or in combination with glucocorticoids.
Collapse
Affiliation(s)
- Dahara Keyse Carvalho Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 40296-710, BA, Brazil; (D.K.C.S.); (L.B.d.C.N.); (C.S.M.)
| | - Laura Beatriz da Cruz Novo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 40296-710, BA, Brazil; (D.K.C.S.); (L.B.d.C.N.); (C.S.M.)
| | - Ivone Maria Ribeiro
- Laboratory of Natural Products Chemistry—PN2, Farmanguinhos, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 22775-903, RJ, Brazil;
| | - Breno Cardim Barreto
- Institute for Innovation in Advanced Health Systems, National Service for Industrial Learning—Integrated Center for Manufacturing and Technology (SENAI CIMATEC), Salvador 41650-010, BA, Brazil; (B.C.B.); (L.C.F.O.)
| | - Luiza Carolina França Opretzka
- Institute for Innovation in Advanced Health Systems, National Service for Industrial Learning—Integrated Center for Manufacturing and Technology (SENAI CIMATEC), Salvador 41650-010, BA, Brazil; (B.C.B.); (L.C.F.O.)
| | - Cássio Santana Meira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 40296-710, BA, Brazil; (D.K.C.S.); (L.B.d.C.N.); (C.S.M.)
- Institute for Innovation in Advanced Health Systems, National Service for Industrial Learning—Integrated Center for Manufacturing and Technology (SENAI CIMATEC), Salvador 41650-010, BA, Brazil; (B.C.B.); (L.C.F.O.)
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 40296-710, BA, Brazil; (D.K.C.S.); (L.B.d.C.N.); (C.S.M.)
- Institute for Innovation in Advanced Health Systems, National Service for Industrial Learning—Integrated Center for Manufacturing and Technology (SENAI CIMATEC), Salvador 41650-010, BA, Brazil; (B.C.B.); (L.C.F.O.)
| |
Collapse
|
50
|
Ward AI, de las Heras JI, Schirmer EC, Fassati A. Memory CD4+ T cells sequentially restructure their 3D genome during stepwise activation. Front Cell Dev Biol 2025; 13:1514627. [PMID: 40018706 PMCID: PMC11866950 DOI: 10.3389/fcell.2025.1514627] [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: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 03/01/2025] Open
Abstract
Background CD4+ T cells are a highly differentiated cell type that maintain enough transcriptomic plasticity to cycle between activated and memory statuses. How the 1D chromatin state and 3D chromatin architecture support this plasticity is under intensive investigation. Methods Here, we wished to test a commercially available in situ Hi-C kit (Arima Genomics Inc.) to establish whether published performance on limiting cell numbers from clonal cell lines copies across to a primary immune cell type. We achieved comparable contact matrices from 50,000, 250,000, and 1,000,000 memory CD4+ T-cell inputs. We generated multiple Hi-C and RNA-seq libraries from the same biological blood donors under three separate conditions: unstimulated fresh ex vivo, IL-2-only stimulated, and T cell receptor (TCR)+CD28+IL-2-stimulated, conferring increasingly stronger activation signals. We wished to capture the magnitude and progression of 3D chromatin shifts and correlate these to expression changes under the two stimulations. Results Although some genome organization changes occurred concomitantly with changes in gene expression, at least as many changes occurred without corresponding changes in expression. Counter to the hypothesis that topologically associated domains (TADs) are largely invariant structures providing a scaffold for dynamic looping contacts between enhancers and promotors, we found that there were at least as many dynamic TAD changes. Stimulation with IL-2 alone triggered many changes in genome organization, and many of these changes were strengthened by additional TCR and CD28 co-receptor stimulation. Conclusions This suggests a stepwise process whereby mCD4+ T cells undergo sequential buildup of 3D architecture induced by distinct or combined stimuli likely to "prime" or "deprime" them for expression responses to subsequent TCR-antigen ligation or additional cytokine stimulation.
Collapse
Affiliation(s)
- Alexander I. Ward
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | | | - Eric C. Schirmer
- Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Ariberto Fassati
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|