1
|
Fan G, Liu Y, Tao L, Wang D, Huang Y, Yang X. Sodium butyrate alleviates colitis by inhibiting mitochondrial ROS mediated macrophage pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167756. [PMID: 40044062 DOI: 10.1016/j.bbadis.2025.167756] [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: 11/20/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease with unclear causes and limited treatment options. Sodium butyrate (NaB), a byproduct of dietary fiber in the intestine, has demonstrated efficacy in treating inflammation. However, the precise anti-inflammatory mechanisms of NaB in colon inflammation remain largely unexplored. This study aims to investigate the effects of NaB on dextran sulfate sodium (DSS)-induced colitis in rats. The findings indicate that oral administration of NaB effectively prevent colitis and reduce levels of serum or colon inflammatory factors. Additionally, NaB demonstrated in vitro inhibition of RAW264.7 inflammation cytokines induced by LPS, along with suppression of the ERK and NF-κB signaling pathway activation. Moreover, NaB mitigated LPS and Nigericin-induced RAW264.7 pyroptosis by reducing indicators of mitochondrial damage, including increased mitochondrial membrane potential (JC-1) levels and decreased Mito-ROS production. NaB increases ZO-1 and Occludin expression in CaCo2 cells by inhibiting RAW264.7 pyroptosis. These results suggest that NaB could be utilized as a therapeutic agent or dietary supplement to alleviate colitis.
Collapse
Affiliation(s)
- Guoqiang Fan
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaxin Liu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Limei Tao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Danping Wang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yizhu Huang
- Singao Xiamen Company, Xiamen 361006, PR China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
2
|
Wei L, Zhu W, Dong C, Kim JK, Ma Y, Denning TL, Kang SM, Wang BZ. Lipid nanoparticles encapsulating both adjuvant and antigen mRNA improve influenza immune cross-protection in mice. Biomaterials 2025; 317:123039. [PMID: 39724768 DOI: 10.1016/j.biomaterials.2024.123039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/26/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
The rapid approval of SARS-CoV-2 mRNA lipid nanoparticle (LNP) vaccines indicates the versatility of mRNA LNPs in an urgent vaccine need. However, the mRNA vaccines do not induce mucosal cellular responses or broad protection against recent variants. To improve cross-protection of mRNA vaccines, here we engineered a pioneered mRNA LNP encapsulating with mRNA constructs encoding cytokine adjuvant and influenza A hemagglutinin (HA) antigen for intradermal vaccination. The adjuvant mRNA encodes a novel fusion cytokine GIFT4 comprising GM-CSF and IL-4. We found that the adjuvanted mRNA LNP vaccine induced high levels of humoral antibodies and systemic T cell responses against heterologous influenza antigens and protected immunized mice against influenza A viral infections. Also, the adjuvanted mRNA LNP vaccine elicited early germinal center reactions in draining lymph nodes and promoted antibody-secreting B cell responses. In addition, we generated another adjuvant mRNA encoding CCL27, which enhanced systemic immune responses. We found the two adjuvant mRNAs both showed effective adjuvanticity in enhancing humoral and cellular responses in mice. Interestingly, intradermal immunizations of GIFT4 or CCL27 mRNA adjuvanted mRNA LNP vaccines induced significant lung tissue-resident T cells. Our findings demonstrate that the cytokine mRNA can be a promising adjuvant flexibly formulated into mRNA LNP vaccines to provoke strong immunity against viral variants.
Collapse
Affiliation(s)
- Lai Wei
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
3
|
Dong Q, Wu W, Zhang R. Mechanistic insights into granulocyte-macrophage colony-stimulating factor in combating fungal infections: Diverse fungal pathogens. Med Mycol 2025; 63:myaf044. [PMID: 40328463 DOI: 10.1093/mmy/myaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/15/2025] [Accepted: 05/03/2025] [Indexed: 05/08/2025] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been used for its immunomodulatory properties to enhance therapeutic outcomes and improve cure rates in fungal infections. However, the mechanisms of GM-CSF action in various fungal infections have not been systematically summarized in current literature, and the reliability and broad effectiveness of clinical data remain uncertain. This review provides a comprehensive analysis of how GM-CSF supports host defense against infections caused by specific fungal pathogens. These pathogens include yeasts (Candida spp., Cryptococcus spp.), filamentous fungi (Aspergillus spp., Mucorales, dematiaceous fungi), and thermally dimorphic fungi (Histoplasma capsulatum, Talaromyces marneffei, Paracoccidioides brasiliensis, and Blastomyces dermatitidis). These insights underscore the potential of GM-CSF as a promising adjunctive therapy in combating challenging fungal infections.
Collapse
Affiliation(s)
- Qi Dong
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Weiwei Wu
- Department of Dermatology, Affiliated Dermatology Hospital of Hainan Medical University, Haikou, Hainan, China
- Department of Dermatology, the Fifth People's Hospital of Hainan Province, Haikou, Hainan, China
| | - Ruijun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
4
|
Zhang F, Jozani KA, Chakravarty A, Lin D, Hollinger A, Rajasekar S, Zhang B. Immune-Infiltrated Cancer Spheroid Model with Vascular Recirculation Reveals Temporally Dependent and Tissue-Specific Macrophage Recruitment. Adv Healthc Mater 2025; 14:e2402946. [PMID: 39962817 PMCID: PMC11973944 DOI: 10.1002/adhm.202402946] [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: 08/07/2024] [Revised: 01/20/2025] [Indexed: 04/08/2025]
Abstract
Immune cell infiltration in tumors has been reported to influence tumor progression and clinical outcomes. Considerable efforts have been made to understand interactions between tumors and the immune system. However, current models are either not comprehensive or limited to short-term studies. Recognizing thedynamic and long-term nature of tumor-immune interactions, an immune-infiltrated cancer spheroid model is developed by continuously perfusing and recirculating immune cells with gravity-driven flow through a tubular blood vessel adjacent to a cancer spheroid. Fibroblasts and pericytes are embedded in the gel matrix to support endothelial cells and enhance the vascular barrier. With continuous monocyte recirculation, monocyte adhesion, transendothelium migration, differentiation, and macrophage recruitment into breast carcinoma and hepatoma spheroids is successfully demonstrated over a week. The macrophage recruitment process is temporally dependent and tissue-specific, leading to the formation of cancer-macrophage heterospheroids. Elevated secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF), which regulates monocyte recruitment and macrophage activation, is observed in the breast carcinoma model. Increased levels of Interleukin 6 (IL-6) and Interleukin 8 (IL-8) are detected, indicating a pro-inflammatory environment associated with tumor progression and metastasis. This platform provides a valuable framework for investigating immune cell infiltration and differentiation within the tumor microenvironment, supporting the advancement of cancer immunotherapies.
Collapse
Affiliation(s)
- Feng Zhang
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Kimia Asadi Jozani
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Anushree Chakravarty
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Dawn Lin
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Andrew Hollinger
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Shravanthi Rajasekar
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Boyang Zhang
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
- The Centre for Discovery in Cancer ResearchMcMaster University1280 Main Street WestHamiltonOntarioL8S 4M1Canada
| |
Collapse
|
5
|
da Silva Fernandes Ribas A, de Godoi KS, Sant’Anna SS, da Rocha MMT, da Silva WD. Release of Cytokines in the Peritoneal Fluid of C57BL/6 Mice After Bothrops jararaca and Bothrops atrox Venom Injection. Toxins (Basel) 2025; 17:164. [PMID: 40278662 PMCID: PMC12030769 DOI: 10.3390/toxins17040164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
The release of cytokines in the peritoneal fluid after stimulation with Bothrops atrox and Bothrops jararaca venoms is a crucial process in the inflammatory response triggered by these venoms. The toxins present in the venoms of snakes from the Bothrops genus induce a complex inflammatory response, which includes the production and release of pro-inflammatory cytokines such as TNF-α, IFN-γ, IL-6, IL-10, IL-1β, chemokines like GM-CSF, MCP-1, and the mast cell degranulation marker MCPT-1. These cytokines play a central role in amplifying inflammation, recruiting leukocytes, and increasing vascular permeability, resulting in edema, pain, and tissue damage at the inoculation site. Peritoneal fluid is commonly used in experimental studies to investigate local inflammatory responses, allowing for the evaluation of the dynamics of inflammatory molecule release. In this study, we used female C57BL/6 mice and observed that Bothrops atrox venom induced a significantly more intense inflammatory response compared to Bothrops jararaca venom. Specifically, Bothrops atrox venom led to a higher release of TNF-α and an increase in MCP-1 levels in peritoneal fluid when compared to Bothrops jararaca venom. These changes resulted in a more pronounced inflammatory condition, with increased leukocyte recruitment in the Bothrops atrox group. Understanding the cytokine profile released in response to these venoms can provide important insights into the pathophysiological mechanisms involved in snakebite accidents and contribute to the development of more effective treatments, such as antivenoms and inflammation modulators.
Collapse
Affiliation(s)
| | - Kemily Stephanie de Godoi
- Immunochemistry Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (A.d.S.F.R.); (K.S.d.G.)
| | | | | | - Wilmar Dias da Silva
- Immunochemistry Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (A.d.S.F.R.); (K.S.d.G.)
| |
Collapse
|
6
|
Robinson GI, Gerasymchuk M, Zanikov T, Gojani EG, Asghari S, Groves A, Haselhorst L, Nandakumar S, Stahl C, Cruz C, Cameron M, Zahoruiko Y, Li D, Rodriguez-Juarez R, Snelling A, Hudson D, Fiselier A, Kovalchuk O, Kovalchuk I. LPS-Induced Liver Inflammation Is Inhibited by Psilocybin and Eugenol in Mice. Pharmaceuticals (Basel) 2025; 18:451. [PMID: 40283890 PMCID: PMC12030523 DOI: 10.3390/ph18040451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Liver inflammatory diseases are a major global health burden and are often exacerbated by inflammation driven by lipopolysaccharides (LPS) through toll-like receptor 4 signaling. This study evaluates the anti-inflammatory effects of psilocybin and eugenol in an LPS-induced liver inflammation model in C57BL/6J mice. Methods: Mice were treated with psilocybin (0.88 mg/kg) and/or eugenol (17.59 mg/kg) either before (pre-treatment) or after (post-treatment) LPS injection. Results: Psilocybin and eugenol, individually and in combination, significantly reduced the LPS-induced mRNA levels of pro-inflammatory cytokines, with post-treatment administration exhibiting stronger effects than pre-treatment. Psilocybin alone displayed the most pronounced anti-inflammatory response, especially for IL-1β, IL-6, and MCP-1, while its combination with eugenol in 1:50 ratio demonstrated similar results, with strongly reduced COX-2 and TNF-α. Histological analysis revealed improved nuclear circularity and reduced inflammatory infiltration in the treatment groups. Eugenol alone showed potential adverse effects, including increased MCP-1 and GM-CSF, but this was mitigated by the co-administration of psilocybin. Conclusions: These findings highlight psilocybin and its combination with eugenol as promising therapies for hepatic inflammation, suggesting their application in treating acute and chronic liver diseases. Future research should explore their long-term effects, the mechanisms underlying their anti-inflammatory actions, and their therapeutic efficacy in humans.
Collapse
Affiliation(s)
- Gregory Ian Robinson
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Marta Gerasymchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Timur Zanikov
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Esmaeel Ghasemi Gojani
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Shima Asghari
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Alyssa Groves
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Lucie Haselhorst
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- Institute for Medical Nutrition Science, Universität zu Lübeck, 23562 Lübeck, Germany
| | - Sanjana Nandakumar
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Cora Stahl
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- Department of Medicine, Medical Sciences, and Nutrition, King’s College, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Ceejay Cruz
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Mackenzie Cameron
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Yeva Zahoruiko
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Dongping Li
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Rocio Rodriguez-Juarez
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Alex Snelling
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Darryl Hudson
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Anna Fiselier
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| |
Collapse
|
7
|
Carvalho JC, Pascoal-Xavier MA, Araújo MG, Martins JP, Teixeira-Carvalho A, Gomes MDS, Amaral LR, Peruhype-Magalhães V, Coelho-dos-Reis JGA, Martins-Filho OA, Araújo MSS. Immune mediators as plasma biomarkers for identifying household contacts and classifying clinical forms and leprosy reactions. Front Immunol 2025; 16:1513060. [PMID: 40109344 PMCID: PMC11920121 DOI: 10.3389/fimmu.2025.1513060] [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: 10/17/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
The present study aimed to evaluate the performance of plasma immune mediators in classifying leprosy patients [L(PB) and L(MB), paucibacillary and multibacillary leprosy, respectively], leprosy reaction patients (T1LR and T2LR, type 1 and type 2 leprosy reaction, respectively), household contacts (HHC), and non-infected (NI) controls. Quantitative measurements of these immune mediators were carried out using high-throughput multiplex microbead array. The results demonstrated that most of the plasma immune mediators were increased in all clinical groups compared with NI controls. Higher frequencies but lower maximum magnitudes of increase (fold change according to NI) were observed for T1LR (63%, 6.1×) and T2LR (63%, 9.7×) compared with HHC (48%, 68.5×), L(PB) (56%, 8.5×), and L(MB) (48%, 37.9×). The bi-dimensional scattering profiles (magnitude order vs. significance) identified a higher number of immune mediators in T2LR (12/27) compared with HHC (8/27), L(PB) (7/27), L(MB) (5/27), and T1LR (5/27). CXCL8 was selected as the parameter with the highest accuracy and significance [area under the receiver operating characteristic curve (AUC) = 0.98, p = 0.0002] in classifying NI vs. HHC. CCL3 (C-C motif chemokine ligand 3) was the single analyte with moderate accuracy and significance (AUC = 0.74, p = 0.0422) in classifying L(PB) vs. L(MB). IL-9 was selected as an attribute with moderate accuracy and significance (AUC = 0.77, p = 0.0041) in classifying T1LR vs. T2LR. Decision tree algorithms confirmed the high accuracy (96%) of CXCL8 in classifying NI vs. HHC. The use of CCL3 followed by IFN-γ classified L(MB) vs. L(PB) with high accuracy (93%). Moreover, the analysis of IL-9 followed by IL-6 and CXCL10 classified T1RL vs. T2RL with high accuracy (96%). In general, combined stepwise algorithms showed enhanced classification accuracy compared with single-attribute analysis. Together, our findings supported the potential use of plasma immune mediators as complementary laboratory biomarkers for the identification of HHC and the classification of distinct clinical forms of leprosy and leprosy reactions.
Collapse
Affiliation(s)
- Jairo Campos Carvalho
- Instituto René Rachou/FIOCRUZ, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Minas Gerais, Brazil
- Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Antônio Pascoal-Xavier
- Instituto René Rachou/FIOCRUZ, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Minas Gerais, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Anatomia Patológica e Medicina Legal, Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Grossi Araújo
- Universidade Federal de Minas Gerais, Serviço de Dermatologia do Hospital das Clínicas, Belo Horizonte, Minas Gerais, Brazil
| | - Júlia Pereira Martins
- Instituto René Rachou/FIOCRUZ, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Minas Gerais, Brazil
| | - Andrea Teixeira-Carvalho
- Instituto René Rachou/FIOCRUZ, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Minas Gerais, Brazil
| | - Matheus de Souza Gomes
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Laurence Rodrigues Amaral
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Vanessa Peruhype-Magalhães
- Instituto René Rachou/FIOCRUZ, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Minas Gerais, Brazil
| | - Jordana Grazziela Alves Coelho-dos-Reis
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Microbiologia, Laboratório de Virologia Básica e Aplicada, Belo Horizonte, Minas Gerais, Brazil
| | - Olindo Assis Martins-Filho
- Instituto René Rachou/FIOCRUZ, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Minas Gerais, Brazil
| | - Márcio Sobreira Silva Araújo
- Instituto René Rachou/FIOCRUZ, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
8
|
Chakraborty C, Bhattacharya M, Das A, Saha A. Regulation of miRNA in Cytokine Storm (CS) of COVID-19 and Other Viral Infection: An Exhaustive Review. Rev Med Virol 2025; 35:e70026. [PMID: 40032584 DOI: 10.1002/rmv.70026] [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: 10/12/2024] [Revised: 01/29/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
In the initial stage of the COVID-19 pandemic, high case fatality was noted. The case fatality during this was associated with the cytokine storm (CS) or cytokine storm syndrome (CSS). Sometimes, virus infections are due to the excessive secretion of pro-inflammatory cytokines, leading to cytokine storms, which might be directed to ARDS, multi-organ failure, and death. However, it was noted that several miRNAs are involved in regulating cytokines during SARS-CoV-2 and other viruses such as IFNs, ILs, GM-CSF, TNF, etc. The article spotlighted several miRNAs involved in regulating cytokines associated with the cytokine storm caused by SARS-CoV-2 and other viruses (influenza virus, MERS-CoV, SARS-CoV, dengue virus). Targeting those miRNAs might help in the discovery of novel therapeutics, considering CS or CSS associated with different virus infections.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | | | - Arpita Das
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | - Abinit Saha
- Deparment of Zoology, J.K. College, Purulia, India
| |
Collapse
|
9
|
He Z, Li F, Liu M, Liao J, Guo C. Porcine Reproductive and Respiratory Syndrome Virus: Challenges and Advances in Vaccine Development. Vaccines (Basel) 2025; 13:260. [PMID: 40266104 PMCID: PMC11945896 DOI: 10.3390/vaccines13030260] [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: 01/06/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 04/24/2025] Open
Abstract
Persistent infection of porcine reproductive and respiratory syndrome virus (PRRSV) significantly hampers both the quantity and quality of pork production in China. Although PRRSV is widely prevalent worldwide, the absence of effective vaccines has made it one of the major pathogens threatening the sustainable development of the global swine industry. Vaccination remains one of the most effective measures for controlling pathogen infections. However, the continuous genetic recombination and mutation of PRRSV demand more comprehensive strategies to address emerging threats, while ensuring the efficacy and safety of vaccines. This review provides an overview of the latest advances in PRRSV vaccine research, highlighting the importance of understanding the unique strengths and limitations of various vaccines in developing effective therapeutic approaches and vaccination strategies. Moreover, the development of adjuvants and antiviral drugs as adjuncts to combat PRRSV infection offers significant potential for enhancing disease control efforts. With the advancement of technologies such as proteolysis-targeting chimera (PROTAC) and mRNA, new avenues for controlling PRRSV and other pathogens are emerging, offering considerable hope. Ultimately, the goal of these vaccine developments is to alleviate the impact of PRRSV on animal health and the profitability of the swine industry.
Collapse
Affiliation(s)
| | | | | | | | - Chunhe Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.H.); (F.L.); (M.L.); (J.L.)
| |
Collapse
|
10
|
Ji P, Grande-Allen KJ, Balaji S, Birla RK, Keswani SG. Shear Stress Conditioning Promotes a Pro-Inflammatory Response in Porcine Endocardial Endothelial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636291. [PMID: 39975174 PMCID: PMC11838557 DOI: 10.1101/2025.02.03.636291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
PURPOSE Discrete subaortic stenosis (DSS) is a congenital heart disease characterized by a narrowing of the passage below the aortic valve in the left ventricular outflow tract (LVOT) [1]. While endocardial endothelial cells (EECs) are known to play a role in DSS, the response of these cells to shear stress is not known. In this study, we hypothesize that the response of EECs to shear stress in the LVOT is a mediator of DSS. METHODS To test this hypothesis, we conditioned porcine EECs to controlled shear stress regimes using cone and plate bioreactors. Subsequently, we quantified the concentration of proinflammatory cytokine in the conditioned media using the Luminex assay. Bulk-RNA sequencing was used to quantify changes in the genotype of the shear stress conditioned EECs. RESULTS The expression of CD31 was knocked down and subsequently, the changes in release of shear stress induced proinflammatory cytokines released by EECs quantified using the Luminex assay. The results of these studies show that the inflammatory cytokines were highly selected in the conditioning medium, and under bioreactor treatment the cell activated the PI3K-AKT and TNF-a signaling, which also triggered the other immune cell responses though Th1, Th2 and Th17 cell differentiation pathways. Furthermore, CD31 was identified as a mediator of the pro-inflammatory response of shear stress conditioned EECs. CONCLUSIONS The studies provide a clear link between shear stress, and the subsequent proinflammatory response of EECs as a mediator of DSS.
Collapse
Affiliation(s)
- Pengfei Ji
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, Texas, USA
- Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, Texas, USA
- Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, Texas, USA
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, Texas, USA
| | | | - Swathi Balaji
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, Texas, USA
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, Texas, USA
| | - Ravi K. Birla
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, Texas, USA
- Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, Texas, USA
- Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, Texas, USA
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, Texas, USA
| | - Sundeep G. Keswani
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, Texas, USA
- Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, Texas, USA
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
11
|
Xie M, Tsai CY, Woo J, Nuritdinov F, Cristaldo M, Odjourian NM, Antilus-Sainte R, Dougher M, Gengenbacher M. BAFF and APRIL immunotherapy following Bacille Calmette-Guérin vaccination enhances protection against pulmonary tuberculosis in mice. Front Immunol 2025; 16:1551183. [PMID: 39981256 PMCID: PMC11839638 DOI: 10.3389/fimmu.2025.1551183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Bacille Calmette-Guérin (BCG), the only tuberculosis vaccine currently in clinical use, provides inadequate long-term protection. Administered at birth, BCG induces broad immune responses against a large number of antigens shared with Mycobacterium tuberculosis (Mtb), but protection wanes over time. We have previously shown that unconventional B cell subsets play a role in tuberculosis control. Methods High-dimensional flow cytometry and multiplex cytokine analysis were employed to investigate the effects of immunotherapy on BCG-vaccinated mice in an Mtb challenge model. Results In this study, we investigate the potential of recombinant cytokines targeting B cells - B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL) - to modulate BCG immunity and improve protection in mice. Both cytokines play overlapping roles in B cell development and peripheral survival. Following subcutaneous BCG vaccination, immunotherapy with BAFF or APRIL resulted in an increased frequency of unconventional B cells potentially transitioning into antibody-producing plasma cells. Concurrently, we observed an increased frequency of central memory T cells, a subset critical for protective immunity. Changes in cellular immune responses were accompanied by reduced pro-inflammatory cytokine profiles and a contraction of the leukocyte population in lungs. Importantly, mice receiving BCG vaccination followed by BAFF or APRIL immunotherapy exhibited superior long-term protection against pulmonary tuberculosis relative to controls that received only BCG. Conclusion In summary, our findings demonstrate that combining BCG vaccination with B cell targeted immunomodulatory therapies can improve long-term protection against pulmonary tuberculosis, highlighting the continued relevance and underutilized potential of BCG as a vaccine platform.
Collapse
Affiliation(s)
- Min Xie
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Chen-Yu Tsai
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Joshua Woo
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Frank Nuritdinov
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Melissa Cristaldo
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Narineh M. Odjourian
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | | | - Maureen Dougher
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Martin Gengenbacher
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| |
Collapse
|
12
|
Uemura K, Katayama KI, Nishioka T, Watanabe H, Yamada G, Inoue N, Asamura S. Dynamics of Immune Cell Infiltration and Fibroblast-Derived IL-33/ST2 Axis Induction in a Mouse Model of Post-Surgical Lymphedema. Int J Mol Sci 2025; 26:1371. [PMID: 39941140 PMCID: PMC11818732 DOI: 10.3390/ijms26031371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Lymphedema is an intractable disease most commonly associated with lymph node dissection for cancer treatment and can lead to a decreased quality of life. Type 2 T helper (Th2) lymphocytes have been shown to be important in the progression of lymphedema. The activation of IL-33 and its receptor, the suppression of tumorigenicity 2 (ST2) signaling pathway, induces the differentiation of Th2 cells, but its involvement in lymphedema remains unclear. In the present study, we analyzed the dynamics of immune cell infiltration, including the IL-33/ST2 axis, in a mouse tail lymphedema model. Neutrophil infiltration was first detected in the lymphedema tissue on postoperative day (POD) 2. Macrophage infiltration increased from POD 2 to 5. The number of CD4+ T cells, including 50% Tregs, gradually increased from POD 14. The mRNA expression of ll13 and Ifng increased on POD 21. The expression of IL-33 was induced in fibroblast nuclei within dermal and subcutaneous tissues from POD 2, and the expression of the Il1rl1 gene encoding ST2 increased from POD 7. We demonstrated the infiltration process from innate to acquired immune cells through the development of a mouse tail lymphedema. The IL-33/ST2 axis was found to be induced during the transition from innate to acquired immunity.
Collapse
Affiliation(s)
- Kazuhisa Uemura
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan; (K.U.); (T.N.); (H.W.); (G.Y.); (S.A.)
- Department of Molecular Genetics, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan;
| | - Kei-ichi Katayama
- Department of Molecular Genetics, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan;
| | - Toshihiko Nishioka
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan; (K.U.); (T.N.); (H.W.); (G.Y.); (S.A.)
| | - Hikaru Watanabe
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan; (K.U.); (T.N.); (H.W.); (G.Y.); (S.A.)
| | - Gen Yamada
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan; (K.U.); (T.N.); (H.W.); (G.Y.); (S.A.)
| | - Norimitsu Inoue
- Department of Molecular Genetics, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan;
| | - Shinichi Asamura
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan; (K.U.); (T.N.); (H.W.); (G.Y.); (S.A.)
| |
Collapse
|
13
|
Kar S, Verma D, Mehrotra S, Prajapati VK. Reconfiguring the immune system to target cancer: Therapies based on T cells, cytokines, and vaccines. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:77-150. [PMID: 39978976 DOI: 10.1016/bs.apcsb.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Over the years, extensive research has been dedicated to performing in-depth analysis of cancer to uncover the intricate details of its nature - including the types of cancer, causative agents, stimulators of disease progression, factors contributing to poor prognosis, and efficient therapies to restrict the metastatic aggressiveness. This chapter highlights the mechanisms through which different arms of the host immune system - namely cytokines, lymphocytes, antigen-presenting cells (APCs) -can be mobilized to eradicate cancer. Most malignant tumors are either poorly immunogenic, or are harbored in a highly immuno-suppressive microenvironment. This is why reinforcing the host's anti-tumor defenses, through infusion of pro-inflammatory cytokines, tumor antigen-loaded APCs, and anti-tumor cytotoxic cells has emerged as a viable treatment option against cancer. The chapter also highlights the ongoing preclinical and clinical studies in different malignancies and the outcome of various therapies. Although these methods are not foolproof, and antigen escape variants can still evade or develop resistance to customized therapies, they achieve disease stabilization in several cases when conventional treatments fail. In many instances, combination therapies involving cytokines, T cells, and vaccinations prove more effective than monotherapies. The limitations of the current therapies are also discussed, along with ongoing modifications aimed at improving efficacy.
Collapse
Affiliation(s)
- Sramona Kar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Divya Verma
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
14
|
Shen YC, Huang YL, Kang YN, Chiu WK, Hoang KD, Wang HJ, Chen C. Effectiveness and safety of talimogene laherparepvec and granulocyte-macrophage colony-stimulating factor for metastatic melanoma: a systematic review and network meta-analysis of randomized controlled trials. Melanoma Res 2025; 35:60-66. [PMID: 39787519 DOI: 10.1097/cmr.0000000000001010] [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: 01/12/2025]
Abstract
Melanoma is an aggressive tumor that is challenging to treat. Talimogene laherparepvec (T-VEC), the first oncolytic virus treatment approved by the US Food and Drug Administration to treat unresectable melanoma, was recently used in recurrent tumors after initial surgery. Our network meta-analysis aimed to compare T-VEC treatment of metastatic melanoma with treatment of granulocyte-macrophage colony-stimulating factor (GM-CSF) and control group. The protocol for this network meta-analysis was retrospectively registered with PROSPERO (CRD42022363321). Three databases, namely Embase, PubMed, and Cochrane Library, were searched until 10 June 2024. The search terms used were a combination of 'metastatic melanoma', 'melanoma', 'T-VEC', 'talimogene laherparepvec', and 'GM-CSF'. Seven studies, with 978 participants receiving T-VEC treatment, 649 participants receiving GM-CSF treatment, and 938 participants constituting the control group, were included in our meta-analysis. For 1-year overall survival (OS), the league table revealed significant differences between the control and T-VEC groups [0.90 (0.83, 0.99)]. The disease-free survival (DFS) over 2 years was also analyzed showing no difference between the groups in DFS in the league table. T-VEC may be a favorable treatment for metastatic melanoma owing to the notable increase in OS. Nevertheless, due to the side effects and limitations, the clinical benefits of T-VEC therapy in metastatic melanoma should be interpreted cautiously. This network meta-analysis demonstrates that T-VEC may be a favorable choice of treatment for metastatic melanoma.
Collapse
Affiliation(s)
- Yu-Chun Shen
- School of Medicine, College of Medicine, Taipei Medical University
| | - Ya-Li Huang
- Department of Public Health, College of Medicine, Taipei Medical University
- Cochrane Taiwan, Taipei Medical University
| | - Yi-No Kang
- Cochrane Taiwan, Taipei Medical University
- Research Center of Big Data and Meta-analysis, Wan Fang Hospital, Taipei Medical University
- Institute of Health Policy and Management, College of Public Health, National Taiwan University
| | - Wen-Kuan Chiu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University
- Division of Plastic Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Khanh Dinh Hoang
- Department of Histopathology, Hai Phong University of Medicine and Pharmacy, Hai Phong, Vietnam
- Evidence-Based Medicine Center, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hsian-Jenn Wang
- Division of Plastic Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chiehfeng Chen
- Department of Public Health, College of Medicine, Taipei Medical University
- Cochrane Taiwan, Taipei Medical University
- Division of Plastic Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Evidence-Based Medicine Center, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
Balkhi S, Bilato G, De Lerma Barbaro A, Orecchia P, Poggi A, Mortara L. Efficacy of Anti-Cancer Immune Responses Elicited Using Tumor-Targeted IL-2 Cytokine and Its Derivatives in Combined Preclinical Therapies. Vaccines (Basel) 2025; 13:69. [PMID: 39852848 PMCID: PMC11768832 DOI: 10.3390/vaccines13010069] [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: 11/25/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Effective cancer therapies must address the tumor microenvironment (TME), a complex network of tumor cells and stromal components, including endothelial, immune, and mesenchymal cells. Durable outcomes require targeting both tumor cells and the TME while minimizing systemic toxicity. Interleukin-2 (IL-2)-based therapies have shown efficacy in cancers such as metastatic melanoma and renal cell carcinoma but are limited by severe side effects. Innovative IL-2-based immunotherapeutic approaches include immunotoxins, such as antibody-drug conjugates, immunocytokines, and antibody-cytokine fusion proteins that enhance tumor-specific delivery. These strategies activate cytotoxic CD8+ T lymphocytes and natural killer (NK) cells, eliciting a potent Th1-mediated anti-tumor response. Modified IL-2 variants with reduced Treg cell activity further improve specificity and reduce immunosuppression. Additionally, IL-2 conjugates with peptides or anti-angiogenic agents offer improved therapeutic profiles. Combining IL-2-based therapies with immune checkpoint inhibitors (ICIs), anti-angiogenic agents, or radiotherapy has demonstrated synergistic potential. Preclinical and clinical studies highlight reduced toxicity and enhanced anti-tumor efficacy, overcoming TME-driven immune suppression. These approaches mitigate the limitations of high-dose soluble IL-2 therapy, promoting immune activation and minimizing adverse effects. This review critically explores advances in IL-2-based therapies, focusing on immunotoxins, immunocytokines, and IL-2 derivatives. Emphasis is placed on their role in combination strategies, showcasing their potential to target the TME and improve clinical outcomes effectively. Also, the use of IL-2 immunocytokines in "in situ" vaccination to relieve the immunosuppression of the TME is discussed.
Collapse
Affiliation(s)
- Sahar Balkhi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.); (L.M.)
| | - Giorgia Bilato
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.); (L.M.)
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20123 Milan, Italy
| | - Andrea De Lerma Barbaro
- Laboratory of Comparative Physiopathology, Department of Biotechnology and Life Sciences, University of Insubria, 20145 Varese, Italy;
| | - Paola Orecchia
- Pathology and Experimental Immunology Operative Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Alessandro Poggi
- SSD Oncologia Molecolare e Angiogenesi, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.); (L.M.)
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20123 Milan, Italy
| |
Collapse
|
16
|
Zhang TH, Chen X, Wei YY, Tang XC, Xu LH, Cui HR, Liu HC, Wang ZX, Chen T, Li CB, Wang JJ. Associations between cytokine levels and cognitive function among individuals at clinical high risk for psychosis. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111166. [PMID: 39383934 DOI: 10.1016/j.pnpbp.2024.111166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
OBJECTIVE To explore the intricate interplay among cytokines, cognitive functioning, and conversion to psychosis in individuals at clinical high-risk (CHR) for psychosis. METHOD We initially enrolled 385 individuals at CHR and 95 healthy controls (HCs). Subsequently, 102 participants at CHR completed the 1-year follow-up assessments, and 47 participants transitioned to psychosis. We assessed the levels of interleukins (IL-1β, IL-2, IL-6, IL-8, IL-10), tumor necrosis factor-α (TNF-α), granulocyte-macrophage colony-stimulating factor (GM-CSF), and vascular endothelial growth factor (VEGF). We comprehensively evaluated cognitive performance across six domains, including speed of processing (SP), attention/vigilance (AV), working memory (WM), verbal learning (VeL), visual learning (ViL), and reasoning and problem-solving (RPS). RESULTS Higher baseline cognitive domain scores were associated with elevated GM-CSF and reduced VEGF levels. In the follow-up analysis, significant time effects were observed for IL-1β and IL-2. We also observed significant interaction effects between specific cognitive domains (AV, WM, VeL, and OCS) and levels of cytokine (GM-CSF, IL-1β, IL-6, and TNF-α). Changes in WM were negatively correlated with changes in TNF-α levels and positively correlated with changes in VEGF levels. Variations in VeL were inversely correlated with changes in GM-CSF and IL-10 levels, whereas changes in RPS were positively associated with changes in GM-CSF and IL-8 levels. CONCLUSIONS Our results revealed intricate associations among cytokine levels, cognitive performance, and psychosis progression.
Collapse
Affiliation(s)
- Tian Hong Zhang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China.
| | - Xing Chen
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China; Department of Psychiatry, Nantong Fourth People's Hospital and Nantong Brain Hospital, NanTong, Jiangsu, China
| | - Yan Yan Wei
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China
| | - Xiao Chen Tang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China
| | - Li Hua Xu
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China
| | - Hui Ru Cui
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China
| | - Hai Chun Liu
- Department of Automation, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zi Xuan Wang
- Shanghai Xinlianxin Psychological Counseling Center, Shanghai, China
| | - Tao Chen
- Big Data Research Lab, University of Waterloo, Ontario, Canada; Labor and Worklife Program, Harvard University, Cambridge, MA, United States
| | - Chun Bo Li
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China
| | - Ji Jun Wang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China; Department of Psychiatry, Nantong Fourth People's Hospital and Nantong Brain Hospital, NanTong, Jiangsu, China; Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Science, Shanghai, PR China; Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, PR China.
| |
Collapse
|
17
|
Miedema J, Cinetto F, Smed-Sörensen A, Spagnolo P. The immunopathogenesis of sarcoidosis. J Autoimmun 2024; 149:103247. [PMID: 38734536 DOI: 10.1016/j.jaut.2024.103247] [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: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Sarcoidosis is a granulomatous multiorgan disease, thought to result from exposure to yet unidentified antigens in genetically susceptible individuals. The exaggerated inflammatory response that leads to granuloma formation is highly complex and involves the innate and adaptive immune system. Consecutive immunological studies using advanced technology have increased our understanding of aberrantly activated immune cells, mediators and pathways that influence the formation, maintenance and resolution of granulomas. Over the years, it has become increasingly clear that disease immunopathogenesis can only be understood if the clinical heterogeneity of sarcoidosis is taken into consideration, along with the distribution of immune cells in peripheral blood and involved organs. Most studies offer an immunological snapshot during disease course, while the cellular composition of both the circulation and tissue microenvironment may change over time. Despite these challenges, novel insights on the role of the immune system are continuously published, thus bringing the field forward. This review highlights current knowledge on the innate and adaptive immune responses involved in sarcoidosis pathogenesis, as well as the pathways involved in non-resolving disease and fibrosis development. Additionally, we describe proposed immunological mechanisms responsible for drug-induced sarcoid like reactions. Although many aspects of disease immunopathogenesis remain to be unraveled, the identification of crucial immune reactions in sarcoidosis may help identify new treatment targets. We therefore also discuss potential therapies and future strategies based on the latest immunological findings.
Collapse
Affiliation(s)
- Jelle Miedema
- Department of Pulmonary Medicine, Center of Expertise for Interstitial Lung Disease, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Italy; Department of Medicine - DIMED, University of Padova, Padova, Italy.
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy.
| |
Collapse
|
18
|
Ma D, Muñoz X, Ojanguren I, Romero-Mesones C, Soler-Segovia D, Varona-Porres D, Cruz MJ. Increased TGFβ1, VEGF and IFN-γ in the Sputum of Severe Asthma Patients With Bronchiectasis. Arch Bronconeumol 2024; 60:682-689. [PMID: 38908944 DOI: 10.1016/j.arbres.2024.05.036] [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: 01/15/2024] [Revised: 05/09/2024] [Accepted: 05/16/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Bronchiectasis is one of the most common comorbidities in severe asthma. However, the mechanisms by which asthma promotes the development and progress of this condition are not well defined. This study aimed to analyze the inflammatory phenotypes and quantify the expression of proinflammatory and remodeling cytokines in asthma patients with and without bronchiectasis. METHODS The study sample comprised individuals with severe asthma and bronchiectasis (group AB, n=55) and a control population of individuals with severe asthma without bronchiectasis (group AC, n=45). Induced sputum samples were obtained and cell types determined by differential cell count. Proinflammatory and bronchial remodeling cytokines (IL-8, neutrophilic elastase, TGFβ1, VEGF, IFN-γ, TNF-α, and GM-CSF) were analyzed by immunoassay in sputum supernatant. RESULTS Neutrophilic inflammation was the primary phenotype in both asthma groups. Higher levels of TGFβ1, VEGF and IFN-γ were observed in asthma patients with bronchiectasis (group AB) than in controls (group AC) (15 vs 24pg/ml, p=0.014; 183 vs 272pg/ml, p=0.048; 0.85 vs 19pg/ml, p<0.001, respectively). Granulocyte-macrophage colony-stimulating factor (GM-CSF) levels were significantly lower in the AB group than in the AC group (1.2 vs 4.4pg/ml, p<0.001). IL-8, neutrophil elastase and TNF-α did not present significant differences between the groups. CONCLUSIONS Raised levels of TGFβ1 and VEGF cytokines may indicate airway remodeling activation in asthma patients with bronchiectasis. The type of inflammation in asthma patients did not differ according to the presence or absence of bronchiectasis.
Collapse
Affiliation(s)
- Donghai Ma
- Servicio de Neumología, Hospital Universitario Vall d́Hebron, Barcelona, Spain
| | - Xavier Muñoz
- Servicio de Neumología, Hospital Universitario Vall d́Hebron, Barcelona, Spain; Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Spain; Departamento de Biología Celular, Fisiología e Inmunología, Universitat Autónoma de Barcelona, Spain.
| | - Iñigo Ojanguren
- Servicio de Neumología, Hospital Universitario Vall d́Hebron, Barcelona, Spain; Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Spain
| | | | - David Soler-Segovia
- Servicio de Neumología, Hospital Universitario Vall d́Hebron, Barcelona, Spain
| | - Diego Varona-Porres
- Servicio de Radiología, Hospital Universitario Vall d́Hebron, Barcelona, Spain
| | - María-Jesús Cruz
- Servicio de Neumología, Hospital Universitario Vall d́Hebron, Barcelona, Spain; Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Spain
| |
Collapse
|
19
|
Wang C, Macintyre AN, Oguin TH, McCarthy KR, Moody MA, Yuan F. Spatiotemporal control of immune responses with nucleic acid cocktail vaccine. ADVANCED THERAPEUTICS 2024; 7:2400263. [PMID: 40248361 PMCID: PMC12002596 DOI: 10.1002/adtp.202400263] [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: 06/18/2024] [Indexed: 04/19/2025]
Abstract
Nucleic acid vaccines play important roles in prevention and treatment of diseases. However, limited immunogenicity remains a major obstacle for DNA vaccine applications in the clinic. To address the issue, the present study investigates a cocktail approach to DNA vaccination. In this proof-of-the-concept study, the cocktail consists of two DNAs encoding viral hemagglutinin (HA) and granulocyte-macrophage colony stimulatory factor (GM-CSF), respectively. Data from the study demonstrate that recruitment and activation of antigen-presenting cells (APCs) can be substantially improved by spatiotemporal regulation of GM-CSF and HA expressions at the site of vaccination. The types of recruited APCs and their phenotypes are also controllable by adjusting the cocktail compositions. Compared to mono-ingredient vaccine, the optimized cocktail vaccine is able to enhance the anti-viral humoral and T cell immune responses. No significant systemic inflammation has been detected after either prime or boost immunization using the cocktail vaccine. Data in the study suggest that the DNA cocktail is a safe, effective, and controllable platform for improving vaccine efficacy.
Collapse
Affiliation(s)
- Chunxi Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Andrew N. Macintyre
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27708, USA
| | - Thomas H. Oguin
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
| | - Kevin R. McCarthy
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA 15261, USA
| | - M. Anthony Moody
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708, USA
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
20
|
Kazakova A, Zhelnov P, Sidorov R, Rogova A, Vasileva O, Ivanov R, Reshetnikov V, Muslimov A. DNA and RNA vaccines against tuberculosis: a scoping review of human and animal studies. Front Immunol 2024; 15:1457327. [PMID: 39421744 PMCID: PMC11483866 DOI: 10.3389/fimmu.2024.1457327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/02/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction To comprehensively identify and provide an overview of in vivo or clinical studies of nucleic acids (NA)-based vaccines against TB we included human or animal studies of NA vaccines for the prevention or treatment of TB and excluded in vitro or in silico research, studies of microorganisms other than M. tuberculosis, reviews, letters, and low-yield reports. Methods We searched PubMed, Scopus, Embase, selected Web of Science and ProQuest databases, Google Scholar, eLIBRARY.RU, PROSPERO, OSF Registries, Cochrane CENTRAL, EU Clinical Trials Register, clinicaltrials.gov, and others through WHO International Clinical Trials Registry Platform Search Portal, AVMA and CABI databases, bioRxiv, medRxiv, and others through OSF Preprint Archive Search. We searched the same sources and Google for vaccine names (GX-70) and scanned reviews for references. Data on antigenic composition, delivery systems, adjuvants, and vaccine efficacy were charted and summarized descriptively. Results A total of 18,157 records were identified, of which 968 were assessed for eligibility. No clinical studies were identified. 365 reports of 345 animal studies were included in the review. 342 (99.1%) studies involved DNA vaccines, and the remaining three focused on mRNA vaccines. 285 (82.6%) studies used single-antigen vaccines, while 48 (13.9%) used multiple antigens or combinations with adjuvants. Only 12 (3.5%) studies involved multiepitope vaccines. The most frequently used antigens were immunodominant secretory antigens (Ag85A, Ag85B, ESAT6), heat shock proteins, and cell wall proteins. Most studies delivered naked plasmid DNA intramuscularly without additional adjuvants. Only 4 of 17 studies comparing NA vaccines to BCG after M. tuberculosis challenge demonstrated superior protection in terms of bacterial load reduction. Some vaccine variants showed better efficacy compared to BCG. Systematic review registration https://osf.io/, identifier F7P9G.
Collapse
Affiliation(s)
- Alisa Kazakova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Pavel Zhelnov
- Zheln, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Roman Sidorov
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Russian Academy of Sciences, Ural Branch, Perm, Russia
| | - Anna Rogova
- Saint-Petersburg State Chemical-Pharmaceutical University, St. Petersburg, Russia
- Laboratory of Nano- and Microencapsulation of Biologically Active Compounds, Peter The Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Olga Vasileva
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Roman Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Vasiliy Reshetnikov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Albert Muslimov
- Saint-Petersburg State Chemical-Pharmaceutical University, St. Petersburg, Russia
| |
Collapse
|
21
|
Tonog G, Yu H, Moon SK, Lee S, Jeong H, Kim HS, Kim KB, Suh HJ, Kim H. Garlic Bioconverted by Bacillus subtilis Stimulates the Intestinal Immune System and Modulates Gut Microbiota Composition. Mol Nutr Food Res 2024; 68:e2400504. [PMID: 39358948 DOI: 10.1002/mnfr.202400504] [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: 07/02/2024] [Revised: 08/30/2024] [Indexed: 10/04/2024]
Abstract
SCOPE This study evaluates the potential of bioconverted garlic ferments (BGFs) to stimulate the intestinal immune system and modulate cecal microbiota composition. METHODS AND RESULTS In vitro, BGF significantly enhances Peyer's patch (PP)-mediated bone marrow cell proliferation and increases the production of interferon-gamma (IFN-γ), granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin (IL)-6, and immunoglobulin A (IgA) but not IL-4, IL-5, and immunoglobulin E (IgE). Oral administration of BGF to C3H/HeN mice for 4 weeks significantly increases the GM-CSF (42.1-45.8 pg mL-1) and IFN-γ (6.5-12.1 pg mL-1) levels in PP cells. BGF also significantly elevates the levels of tumor necrosis factor-alpha (TNF-α, 165.0-236.3 pg mg-1), GM-CSF (2.4-3.0 ng mg-1), and IFN-γ (1.5-3.2 ng mg-1) in the small intestinal fluid, and TNF-α (2.2-3.1 pg mL-1) and IFN-γ (10.3-0.21.5 pg mL-1) in the mouse serum. Cecal microbial analysis reveals that BGF increases Bacteroidota and Verrucomicrobiota and decreases Actinobacteria and Bacillota at the phylum level in mice. At the genus level, BGF significantly increases the abundance of Fusimonas (250 mg kg-1 BW-1 day-1), Bacteroides (125 and 250 mg kg-1 BW-1 day-1), and Akkermansia (125 mg kg-1 BW-1 day-1) and decreases that of Bifidobacterium (62.5 and 250 mg kg-1 BW-1 day-1) and Limosilactobacillus (125 and 250 mg kg-1 BW-1 day-1). CONCLUSION This study provides the first evidence of BGF's ability to modulate the intestinal immune system and gut microbiota, supporting its potential as a novel functional material to enhance gut immunity.
Collapse
Affiliation(s)
- Genevieve Tonog
- Department of Food and Nutrition, Chung-Ang University, Anseong, 17546, South Korea
| | - Hyeonjun Yu
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, South Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong, 17546, South Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong, 17546, South Korea
| | | | | | | | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, South Korea
| | - Hoon Kim
- Department of Food and Nutrition, Chung-Ang University, Anseong, 17546, South Korea
| |
Collapse
|
22
|
Zhang Y, Deng Y, Zhai Y, Li Y, Li Y, Li J, Gu Y, Li S. A bispecific nanosystem activates endogenous natural killer cells in the bone marrow for haematologic malignancies therapy. NATURE NANOTECHNOLOGY 2024; 19:1558-1568. [PMID: 39043825 DOI: 10.1038/s41565-024-01736-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 05/20/2024] [Indexed: 07/25/2024]
Abstract
Haematologic malignancies commonly arise from the bone marrow lesion, yet there are currently no effective targeted therapies against tumour cells in this location. Here we constructed a bone-marrow-targeting nanosystem, CSF@E-Hn, which is based on haematopoietic-stem-cell-derived nanovesicles adorned with gripper ligands (aPD-L1 and aNKG2D) and encapsulated with colony-stimulating factor (CSF) for the treatment of haematologic malignancies. CSF@E-Hn targets the bone marrow and, thanks to the gripper ligands, pulls together tumour cells and natural killer cells, activating the latter for specific tumour cell targeting and elimination. The therapeutic efficacy was validated in mice bearing acute myeloid leukaemia and multiple myeloma. The comprehensive assessment of the post-treatment bone marrow microenvironment revealed that the integration of CSF into a bone-marrow-targeted nanosystem promoted haematopoietic stem cell differentiation, boosted memory T cell generation and maintained bone homoeostasis, with long-term prevention of relapse. Our nanosystem represents a promising strategy for the treatment of haematologic malignancies.
Collapse
MESH Headings
- Animals
- Mice
- Killer Cells, Natural/immunology
- Killer Cells, Natural/drug effects
- Bone Marrow/drug effects
- Bone Marrow/pathology
- Humans
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/drug therapy
- Hematologic Neoplasms/pathology
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Nanoparticles/chemistry
- Hematopoietic Stem Cells/drug effects
- Multiple Myeloma/drug therapy
- Multiple Myeloma/pathology
- Multiple Myeloma/immunology
- Female
Collapse
Affiliation(s)
- Yanqin Zhang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Yanfang Deng
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Yuewen Zhai
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Yu Li
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Yuting Li
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Juequan Li
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China.
| | - Siwen Li
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
23
|
Bai Z, Feng B, McClory SE, de Oliveira BC, Diorio C, Gregoire C, Tao B, Yang L, Zhao Z, Peng L, Sferruzza G, Zhou L, Zhou X, Kerr J, Baysoy A, Su G, Yang M, Camara PG, Chen S, Tang L, June CH, Melenhorst JJ, Grupp SA, Fan R. Single-cell CAR T atlas reveals type 2 function in 8-year leukaemia remission. Nature 2024; 634:702-711. [PMID: 39322664 PMCID: PMC11485231 DOI: 10.1038/s41586-024-07762-w] [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: 05/28/2023] [Accepted: 06/27/2024] [Indexed: 09/27/2024]
Abstract
Despite a high response rate in chimeric antigen receptor (CAR) T cell therapy for acute lymphocytic leukaemia (ALL)1-3, approximately 50% of patients relapse within the first year4-6, representing an urgent question to address in the next stage of cellular immunotherapy. Here, to investigate the molecular determinants of ultralong CAR T cell persistence, we obtained a single-cell multi-omics atlas from 695,819 pre-infusion CAR T cells at the basal level or after CAR-specific stimulation from 82 paediatric patients with ALL enrolled in the first two CAR T ALL clinical trials and 6 healthy donors. We identified that elevated type 2 functionality in CAR T infusion products is significantly associated with patients maintaining a median B cell aplasia duration of 8.4 years. Analysis of ligand-receptor interactions revealed that type 2 cells regulate a dysfunctional subset to maintain whole-population homeostasis, and the addition of IL-4 during antigen-specific activation alleviates CAR T cell dysfunction while enhancing fitness at both transcriptomic and epigenomic levels. Serial proteomic profiling of sera after treatment revealed a higher level of circulating type 2 cytokines in 5-year or 8-year relapse-free responders. In a leukaemic mouse model, type 2high CAR T cell products demonstrated superior expansion and antitumour activity, particularly after leukaemia rechallenge. Restoring antitumour efficacy in type 2low CAR T cells was attainable by enhancing their type 2 functionality, either through incorporating IL-4 into the manufacturing process or by priming manufactured CAR T products with IL-4 before infusion. Our findings provide insights into the mediators of durable CAR T therapy response and suggest potential therapeutic strategies to sustain long-term remission by boosting type 2 functionality in CAR T cells.
Collapse
Affiliation(s)
- Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Bing Feng
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland
| | - Susan E McClory
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Caroline Diorio
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Céline Gregoire
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bo Tao
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Luojia Yang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Ziran Zhao
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Giacomo Sferruzza
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Liqun Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaolei Zhou
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland
| | - Jessica Kerr
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alev Baysoy
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Graham Su
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Mingyu Yang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Pablo G Camara
- Department of Genetics and Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland.
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Stephan A Grupp
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
- Human and Translational Immunology, Yale University School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
24
|
Zong YH, Cao JF, Zhao Y, Gao M, Chen WL, Wu M, Xu X, Xu ZY, Zhang XQ, Tang JZ, Liu Y, Hu XS, Wang SQ, Zhang X. Mechanism of Lian Hua Qing Wen capsules regulates the inflammatory response caused by M 1 macrophage based on cellular experiments and computer simulations. Acta Trop 2024; 257:107320. [PMID: 39002739 DOI: 10.1016/j.actatropica.2024.107320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE The polarization of macrophages with the resulting inflammatory response play a crucial part in tissue and organ damage due to inflammatory. Study has proved Lian Hua Qing Wen capsules (LHQW) can reduce activation of inflammatory response and damage of tissue derived from the inflammatory reactions. However, the mechanism of LHQW regulates the macrophage-induced inflammatory response is unclear. Therefore, we investigated the mechanism of LHQW regulated the inflammatory response of M1 macrophages by cellular experiments and computer simulations. METHODS This study has analysed the targets and mechanisms of macrophage regulating inflammatory response at gene and protein levels through bioinformatics. The monomeric components of LHQW were analyzed by High Performance Liquid Chromatography (HPLC). We established the in vitro cell model by M1 macrophages (Induction of THP-1 cells into M1 macrophages). RT-qPCR and immunofluorescence were used to detect changes in gene and protein levels of key targets after LHQW treatment. Computer simulations were utilized to verify the binding stability of monomeric components and protein targets. RESULTS Macrophages had 140,690 gene targets, inflammatory response had 12,192 gene targets, intersection gene targets were 11,772. Key monomeric components (including: Pinocembrin, Fargesone-A, Nodakenin and Bowdichione) of LHQW were screened by HPLC. The results of cellular experiments indicated that LHQW could significantly reduce the mRNA expression of CCR5, CSF2, IFNG and TNF, thereby alleviating the inflammatory response caused by M1 macrophage. The computer simulations further validated the binding stability and conformation of key monomeric components and key protein targets, and IFNG/Nodakenin was able to form the most stable binding conformation for its action. CONCLUSION In this study, the mechanism of LHQW inhibits the polarization of macrophages and the resulting inflammatory response was investigated by computer simulations and cellular experiments. We found that LHQW may not only reduce cell damage and death by acting on TNF and CCR5, but also inhibit the immune recognition process and inflammatory response by regulating CSF2 and IFNG to prevent polarization of macrophages. Therefore, these results suggested that LHQW may act through multiple targets to inhibit the polarization of macrophages and the resulting inflammatory response.
Collapse
Affiliation(s)
| | - Jun-Feng Cao
- College of Medicine, Southwest Jiaotong University, Chengdu, PR China
| | | | - Miao Gao
- Chengdu Medical College, Chengdu, PR China
| | | | - Mei Wu
- Chengdu Medical College, Chengdu, PR China
| | - Xiang Xu
- Chengdu Medical College, Chengdu, PR China
| | | | | | | | - Yulin Liu
- Chengdu Medical College, Chengdu, PR China
| | | | | | - Xiao Zhang
- Chengdu Medical College, Chengdu, PR China.
| |
Collapse
|
25
|
Ni J, Wang X, Wu L, Ai X, Chu Q, Han C, Dong X, Zhou Y, Pang Y, Zhu Z. Sintilimab in combination with stereotactic body radiotherapy and granulocyte-macrophage colony-stimulating factor in metastatic non-small cell lung cancer: The multicenter SWORD phase 2 trial. Nat Commun 2024; 15:7242. [PMID: 39174542 PMCID: PMC11341907 DOI: 10.1038/s41467-024-51807-7] [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: 03/03/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024] Open
Abstract
This single-arm, multicenter, phase 2 trial (NCT04106180) investigated the triple combination of sintilimab (anti-PD1 antibody), stereotactic body radiotherapy (SBRT) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in metastatic non-small cell lung cancer (NSCLC). With a median follow-up of 32.1 months, 18 (36.7%, 90% CI 25.3%-49.5%) of the 49 evaluable patients had an objective response, meeting the primary endpoint. Secondary endpoints included out-of-field (abscopal) response rate (ASR), progression-free survival (PFS), overall survival (OS), and treatment-related adverse events (TRAEs). The ASR was 30.6% (95% CI 18.3%-45.4%). The median PFS and OS were 5.9 (95% CI 2.5-9.3) and 18.4 (95% CI 9.7-27.1) months, respectively. Any grade and grade 3 TRAEs occurred in 44 (86.3%) and 6 (11.8%) patients, without grade 4-5 TRAEs. Moreover, in pre-specified biomarker analyses, SBRT-induced increase of follicular helper T cells (Tfh) in unirradiated tumor lesions and patient's blood, as well as of circulating IL-21 levels, was found associated with improved prognosis. Taken together, the triple combination therapy was well tolerated with promising efficacy and Tfh may play a critical role in SBRT-triggered anti-tumor immunity in metastatic NSCLC.
Collapse
Affiliation(s)
- Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaofei Wang
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Lin Wu
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinghao Ai
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengbo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yechun Pang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Thoracic Oncology, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Zhu Y, Zhou L, Mo L, Hong C, Pan L, Lin J, Qi Y, Tan S, Qian M, Hu T, Zhao Y, Qiu H, Lin P, Ma X, Yang Q. Plasmodium yoelii Infection Enhances the Expansion of Myeloid-Derived Suppressor Cells via JAK/STAT3 Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:170-186. [PMID: 38819229 DOI: 10.4049/jimmunol.2300541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs), the negative immune regulators, have been demonstrated to be involved in immune responses to a variety of pathological conditions, such as tumors, chronic inflammation, and infectious diseases. However, the roles and mechanisms underlying the expansion of MDSCs in malaria remain unclear. In this study, the phenotypic and functional characteristics of splenic MDSCs during Plasmodium yoelii NSM infection are described. Furthermore, we provide compelling evidence that the sera from P. yoelii-infected C57BL/6 mice containing excess IL-6 and granulocyte-macrophage colony-stimulating factor promote the accumulation of MDSCs by inducing Bcl2 expression. Serum-induced MDSCs exert more potent suppressive effects on T cell responses than control MDSCs within both in vivo P. yoelii infection and in vitro serum-treated bone marrow cells experiments. Serum treatment increases the MDSC inhibitory effect, which is dependent on Arg1 expression. Moreover, mechanistic studies reveal that the serum effects are mediated by JAK/STAT3 signaling. By inhibiting STAT3 phosphorylation with the JAK inhibitor JSI-124, effects of serum on MDSCs are almost eliminated. In vivo depletion of MDSCs with anti-Gr-1 or 5-fluorouracil significantly reduces the parasitemia and promotes Th1 immune response in P. yoelii-infected C57BL/6 mice by upregulating IFN-γ expression. In summary, this study indicates that P. yoelii infection facilitates the accumulation and function of MDSCs by upregulating the expression of Bcl2 and Arg1 via JAK/STAT3 signaling pathway in vivo and in vitro. Manipulating the JAK/STAT3 signaling pathway or depleting MDSCs could be promising therapeutic interventions to treat malaria.
Collapse
Affiliation(s)
- Yiqiang Zhu
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Lu Zhou
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lengshan Mo
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Cansheng Hong
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lingxia Pan
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jie Lin
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yanwei Qi
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Simin Tan
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Manhongtian Qian
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tengfei Hu
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yi Zhao
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huaina Qiu
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Peibin Lin
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Xiancai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Quan Yang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
27
|
Ahn MJ, Kim EH, Choi Y, Chae CH, Kim P, Kim SH. Novel hematopoietic progenitor kinase 1 inhibitor KHK-6 enhances T-cell activation. PLoS One 2024; 19:e0305261. [PMID: 38923962 PMCID: PMC11207149 DOI: 10.1371/journal.pone.0305261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Inhibiting the functional role of negative regulators in immune cells is an effective approach for developing immunotherapies. The serine/threonine kinase hematopoietic progenitor kinase 1 (HPK1) involved in the T-cell receptor signaling pathway attenuates T-cell activation by inducing the degradation of SLP-76 through its phosphorylation at Ser-376, reducing the immune response. Interestingly, several studies have shown that the genetic ablation or pharmacological inhibition of HPK1 kinase activity improves the immune response to cancers by enhancing T-cell activation and cytokine production; therefore, HPK1 could be a promising druggable target for T-cell-based cancer immunotherapy. To increase the immune response against cancer cells, we designed and synthesized KHK-6 and evaluated its cellular activity to inhibit HPK1 and enhance T-cell activation. KHK-6 inhibited HPK1 kinase activity with an IC50 value of 20 nM and CD3/CD28-induced phosphorylation of SLP-76 at Ser-376 Moreover, KHK-6 significantly enhanced CD3/CD28-induced production of cytokines; proportion of CD4+ and CD8+ T cells that expressed CD69, CD25, and HLA-DR markers; and T-cell-mediated killing activity of SKOV3 and A549 cells. In conclusion, KHK-6 is a novel ATP-competitive HPK1 inhibitor that blocks the phosphorylation of HPK1 downstream of SLP-76, enhancing the functional activation of T cells. In summary, our study showed the usefulness of KHK-6 in the drug discovery for the HPK1-inhibiting immunotherapy.
Collapse
Affiliation(s)
- Min Jeong Ahn
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, Republic of Korea
| | - Eun Hye Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Yunha Choi
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Medicinal Chemistry & Pharmacology, University of Science and Technology, Daejeon, Republic of Korea
| | - Chong Hak Chae
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Pilho Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Medicinal Chemistry & Pharmacology, University of Science and Technology, Daejeon, Republic of Korea
| | - Seong Hwan Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
28
|
Wong JPH, Blazev R, Ng YK, Goodman CA, Montgomery MK, Watt KI, Carl CS, Watt MJ, Voldstedlund CT, Richter EA, Crouch PJ, Steyn FJ, Ngo ST, Parker BL. Characterization of the skeletal muscle arginine methylome in health and disease reveals remodeling in amyotrophic lateral sclerosis. FASEB J 2024; 38:e23647. [PMID: 38787599 DOI: 10.1096/fj.202400045r] [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: 01/08/2024] [Revised: 04/04/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Arginine methylation is a protein posttranslational modification important for the development of skeletal muscle mass and function. Despite this, our understanding of the regulation of arginine methylation under settings of health and disease remains largely undefined. Here, we investigated the regulation of arginine methylation in skeletal muscles in response to exercise and hypertrophic growth, and in diseases involving metabolic dysfunction and atrophy. We report a limited regulation of arginine methylation under physiological settings that promote muscle health, such as during growth and acute exercise, nor in disease models of insulin resistance. In contrast, we saw a significant remodeling of asymmetric dimethylation in models of atrophy characterized by the loss of innervation, including in muscle biopsies from patients with myotrophic lateral sclerosis (ALS). Mass spectrometry-based quantification of the proteome and asymmetric arginine dimethylome of skeletal muscle from individuals with ALS revealed the largest compendium of protein changes with the identification of 793 regulated proteins, and novel site-specific changes in asymmetric dimethyl arginine (aDMA) of key sarcomeric and cytoskeletal proteins. Finally, we show that in vivo overexpression of PRMT1 and aDMA resulted in increased fatigue resistance and functional recovery in mice. Our study provides evidence for asymmetric dimethylation as a regulator of muscle pathophysiology and presents a valuable proteomics resource and rationale for numerous methylated and nonmethylated proteins, including PRMT1, to be pursued for therapeutic development in ALS.
Collapse
Affiliation(s)
- Julian P H Wong
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ronnie Blazev
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Yaan-Kit Ng
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Craig A Goodman
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kevin I Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, Victoria, Australia
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Christian S Carl
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, The University of Copenhagen, Copenhagen, Denmark
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christian T Voldstedlund
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, The University of Copenhagen, Copenhagen, Denmark
| | - Erik A Richter
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, The University of Copenhagen, Copenhagen, Denmark
| | - Peter J Crouch
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Frederik J Steyn
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Shyuan T Ngo
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Sang L, Gong X, Huang Y, Zhang L, Sun J. Immunotherapeutic implications on targeting the cytokines produced in rhinovirus-induced immunoreactions. FRONTIERS IN ALLERGY 2024; 5:1427762. [PMID: 38859875 PMCID: PMC11163110 DOI: 10.3389/falgy.2024.1427762] [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: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
Rhinovirus is a widespread virus associated with several respiratory diseases, especially asthma exacerbation. Currently, there are no accurate therapies for rhinovirus. Encouragingly, it is found that during rhinovirus-induced immunoreactions the levels of certain cytokines in patients' serum will alter. These cytokines may have pivotal pro-inflammatory or anti-inflammatory effects via their specific mechanisms. Thus far, studies have shown that inhibitions of cytokines such as IL-1, IL-4, IL-5, IL-6, IL-13, IL-18, IL-25, and IL-33 may attenuate rhinovirus-induced immunoreactions, thereby relieving rhinovirus infection. Furthermore, such therapeutics for rhinovirus infection can be applied to viruses of other species, with certain practicability.
Collapse
Affiliation(s)
- Le Sang
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Xia Gong
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Yunlei Huang
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Linling Zhang
- Department of Respiratory Medicine, Shaoxing People’s Hospital, Shaoxing City, Zhejiang Province, China
| | - Jian Sun
- Department of Respiratory Medicine, Shaoxing People’s Hospital, Shaoxing City, Zhejiang Province, China
| |
Collapse
|
30
|
Kan AKC, Tang WT, Li PH. Helper T cell subsets: Development, function and clinical role in hypersensitivity reactions in the modern perspective. Heliyon 2024; 10:e30553. [PMID: 38726130 PMCID: PMC11079302 DOI: 10.1016/j.heliyon.2024.e30553] [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: 01/14/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Helper T cells are traditionally classified into T helper 1 (TH1) and T helper 2 (TH2). The more recent discoveries of T helper 17 (TH17), follicular helper T cells (TFH) and regulatory T cells (Treg) enhanced our understanding on the mechanisms of immune function and hypersensitivity reactions, which shaped the modern perspective on the function and role of these different subsets of helper T cells in hypersensitivity reactions. Each subset of helper T cells has characteristic roles in different types of hypersensitivity reactions, hence giving rise to the respective characteristic clinical manifestations. The roles of helper T cells in allergic contact dermatitis (TH1-mediated), drug rash with eosinophilia and systemic symptoms (DRESS) syndrome (TH2-mediated), and acute generalised exanthematous pustulosis (AGEP) (TH17-mediated) are summarised in this article, demonstrating the correlation between the type of helper T cell involved and the clinical features. TFH plays crucial roles in antibody class-switch recombination; they may be implicated in antibody-mediated hypersensitivity reactions, but further research is warranted to delineate their exact pathogenic roles. The helper T cell subsets and their specific cytokine profiles implicated in different hypersensitivity reactions could be potential treatment targets by biologics, but more clinical trials are warranted to establish their clinical effectiveness.
Collapse
Affiliation(s)
- Andy Ka Chun Kan
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region of China
| | - Wang Tik Tang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region of China
| | - Philip H. Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region of China
| |
Collapse
|
31
|
Lee YS, Bang YJ, Yoo S, Park SI, Park HJ, Kwak HW, Bae SH, Park HJ, Kim JY, Youn SB, Roh G, Lee S, Kwon SP, Bang EK, Keum G, Nam JH, Hong SH. Analysis of the Immunostimulatory Effects of Cytokine-Expressing Internal Ribosome Entry Site-Based RNA Adjuvants and Their Applications. J Infect Dis 2024; 229:1408-1418. [PMID: 37711050 DOI: 10.1093/infdis/jiad392] [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: 04/28/2023] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023] Open
Abstract
Developing new adjuvants that can effectively induce humoral and cellular immune responses while broadening the immune response is of great value. In this study, we aimed to develop single-stranded RNA adjuvants expressing (1) granulocyte monocyte colony-stimulating factor or (2) interleukin 18 based on the encephalomyocarditis virus internal ribosome entry site; we also tested their efficacy in combination with ovalbumin or inactivated influenza vaccines. Notably, cytokine-expressing RNA adjuvants increased the expression of antigen-presenting cell activation markers in mice. Specifically, when combined with ovalbumin, RNA adjuvants expressing granulocyte monocyte colony-stimulating factor increased CD4+ T-cell responses, while those expressing interleukin 18 increased CD8+ T-cell responses. Cytokine-expressing RNA adjuvants further increased the frequency of polyclonal T cells with the influenza vaccine and reduced the clinical illness scores and weight loss of mice after viral challenge. Collectively, our results suggest that cytokine-expressing RNA adjuvants can be applied to protein-based or inactivated vaccines to increase their efficacy.
Collapse
Affiliation(s)
- Yu-Sun Lee
- Department of Biotechnology
- BK21 FOUR Department of Biotechnology, The Catholic University of Korea, Bucheon
| | - Yoo-Jin Bang
- Department of Biotechnology
- Central Research Institute, SML Biopharm, Gwangmyeong
| | - Soyeon Yoo
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul
| | - Sang-In Park
- Central Research Institute, SML Biopharm, Gwangmyeong
| | - Hyo-Jung Park
- Department of Biotechnology
- BK21 FOUR Department of Biotechnology, The Catholic University of Korea, Bucheon
| | - Hye Won Kwak
- Central Research Institute, SML Biopharm, Gwangmyeong
| | - Seo-Hyeon Bae
- Department of Biotechnology
- BK21 FOUR Department of Biotechnology, The Catholic University of Korea, Bucheon
| | | | - Jae-Yong Kim
- Department of Biotechnology
- Central Research Institute, SML Biopharm, Gwangmyeong
| | - Sue-Bean Youn
- Department of Biotechnology
- BK21 FOUR Department of Biotechnology, The Catholic University of Korea, Bucheon
| | - Gahyun Roh
- Department of Biotechnology
- BK21 FOUR Department of Biotechnology, The Catholic University of Korea, Bucheon
| | - Seonghyun Lee
- Department of Biotechnology
- BK21 FOUR Department of Biotechnology, The Catholic University of Korea, Bucheon
| | - Sung Pil Kwon
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul
| | - Eun-Kyoung Bang
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul
| | - Gyochang Keum
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul
| | - Jae-Hwan Nam
- BK21 FOUR Department of Biotechnology, The Catholic University of Korea, Bucheon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon
| | - So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
32
|
Yatomi M, Akasaka K, Sato S, Chida M, Kanbe M, Sawada H, Yokota I, Wakamatsu I, Muto S, Sato M, Yamaguchi K, Miura Y, Tsurumaki H, Sakurai R, Hara K, Koga Y, Sunaga N, Yamakawa H, Matsushima H, Yamazaki S, Endo Y, Motegi SI, Hisada T, Maeno T. A case of autoimmune pulmonary alveolar proteinosis during the course of treatment of rapidly progressive interstitial pneumonia associated with anti-MDA5 antibody-positive dermatomyositis. BMC Pulm Med 2024; 24:170. [PMID: 38589870 PMCID: PMC11003183 DOI: 10.1186/s12890-024-02989-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/01/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Autoimmune pulmonary alveolar proteinosis (APAP) is a diffuse lung disease that causes abnormal accumulation of lipoproteins in the alveoli; however, its pathogenesis remains unclear. Recently, APAP cases have been reported during the course of dermatomyositis. The combination of these two diseases may be coincidental; however, it may have been overlooked because differentiating APAP from a flare-up of interstitial pneumonia associated with dermatomyositis is challenging. This didactic case demonstrates the need for early APAP scrutiny. CASE PRESENTATION A 50-year-old woman was diagnosed with anti-melanoma differentiation-associated gene 5 (anti-MDA5) antibody-positive dermatitis and interstitial pneumonia in April 2021. The patient was treated with corticosteroids, tacrolimus, and cyclophosphamide pulse therapy for interstitial pneumonia complicated by MDA5 antibody-positive dermatitis, which improved the symptoms and interstitial pneumonia. Eight months after the start of treatment, a new interstitial shadow appeared that worsened. Therefore, three additional courses of cyclophosphamide pulse therapy were administered; however, the respiratory symptoms and interstitial shadows did not improve. Respiratory failure progressed, and 14 months after treatment initiation, bronchoscopy revealed turbid alveolar lavage fluid, numerous foamy macrophages, and numerous periodic acid-Schiff-positive unstructured materials. Blood test results revealed high anti-granulocyte-macrophage colony-stimulating factor (GM-CSF) antibody levels, leading to a diagnosis of APAP. The patient underwent whole-lung lavage, and the respiratory disturbance promptly improved. Anti-GM-CSF antibodies were measured from the cryopreserved serum samples collected at the time of diagnosis of anti-MDA5 antibody-positive dermatitis, and 10 months later, both values were significantly higher than normal. CONCLUSIONS This is the first report of anti-MDA5 antibody-positive dermatomyositis complicated by interstitial pneumonia with APAP, which may develop during immunosuppressive therapy and be misdiagnosed as a re-exacerbation of interstitial pneumonia. In anti-MDA5 antibody-positive dermatomyositis, APAP comorbidity may have been overlooked, and early evaluation with bronchoalveolar lavage fluid and anti-GM-CSF antibody measurements should be considered, keeping the development of APAP in mind.
Collapse
Affiliation(s)
- Masakiyo Yatomi
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan.
| | - Keiichi Akasaka
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5, Shintoshin, Chuo-Ku, Saitama, 330-8553, Japan
| | - Shintaro Sato
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5, Shintoshin, Chuo-Ku, Saitama, 330-8553, Japan
| | - Mizuki Chida
- Department of Dermatology, Gunma University Graduate School of Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Mio Kanbe
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Hiru Sawada
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Itaru Yokota
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Ikuo Wakamatsu
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Sohei Muto
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Mari Sato
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Kochi Yamaguchi
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Yosuke Miura
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Hiroaki Tsurumaki
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Reiko Sakurai
- Oncology Center, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371-, 8511, Japan
| | - Kenichiro Hara
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Yasuhiko Koga
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Noriaki Sunaga
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Hideaki Yamakawa
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5, Shintoshin, Chuo-Ku, Saitama, 330-8553, Japan
| | - Hidekazu Matsushima
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5, Shintoshin, Chuo-Ku, Saitama, 330-8553, Japan
| | - Sahori Yamazaki
- Department of Dermatology, Gunma University Graduate School of Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Yukie Endo
- Department of Dermatology, Gunma University Graduate School of Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Sei-Ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| | - Takeshi Hisada
- Gunma University Graduate School of Health Sciences, 3-39-22 Showa-Machi, Maebashi, Gunma, 371-8514, Japan
| | - Toshitaka Maeno
- Division of Allergy and Respiratory Medicine, Integrative Center of Internal Medicine, Gunma University Hospital, 3-39-15 Showa-Machi, Maebashi, Gunma, 371- 8511, Japan
| |
Collapse
|
33
|
Lum FM, Chan YH, Teo TH, Becht E, Amrun SN, Teng KW, Hartimath SV, Yeo NK, Yee WX, Ang N, Torres-Ruesta AM, Fong SW, Goggi JL, Newell EW, Renia L, Carissimo G, Ng LF. Crosstalk between CD64 +MHCII + macrophages and CD4 + T cells drives joint pathology during chikungunya. EMBO Mol Med 2024; 16:641-663. [PMID: 38332201 PMCID: PMC10940729 DOI: 10.1038/s44321-024-00028-y] [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: 07/26/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
Communications between immune cells are essential to ensure appropriate coordination of their activities. Here, we observed the infiltration of activated macrophages into the joint-footpads of chikungunya virus (CHIKV)-infected animals. Large numbers of CD64+MHCII+ and CD64+MHCII- macrophages were present in the joint-footpad, preceded by the recruitment of their CD11b+Ly6C+ inflammatory monocyte precursors. Recruitment and differentiation of these myeloid subsets were dependent on CD4+ T cells and GM-CSF. Transcriptomic and gene ontology analyses of CD64+MHCII+ and CD64+MHCII- macrophages revealed 89 differentially expressed genes, including genes involved in T cell proliferation and differentiation pathways. Depletion of phagocytes, including CD64+MHCII+ macrophages, from CHIKV-infected mice reduced disease pathology, demonstrating that these cells play a pro-inflammatory role in CHIKV infection. Together, these results highlight the synergistic dynamics of immune cell crosstalk in driving CHIKV immunopathogenesis. This study provides new insights in the disease mechanism and offers opportunities for development of novel anti-CHIKV therapeutics.
Collapse
Affiliation(s)
- Fok-Moon Lum
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore.
| | - Yi-Hao Chan
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Teck-Hui Teo
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Etienne Becht
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, 138648, Singapore
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Siti Naqiah Amrun
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Karen Ww Teng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Siddesh V Hartimath
- Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Nicholas Kw Yeo
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Wearn-Xin Yee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Nicholas Ang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Anthony M Torres-Ruesta
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Siew-Wai Fong
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Julian L Goggi
- Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, 138648, Singapore
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, 138648, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Guillaume Carissimo
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore
| | - Lisa Fp Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore, 138648, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, L69 7BE, UK.
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 7ZX, UK.
| |
Collapse
|
34
|
Dudek P, Talar-Wojnarowska R. Current Approach to Risk Factors and Biomarkers of Intestinal Fibrosis in Inflammatory Bowel Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:305. [PMID: 38399592 PMCID: PMC10889938 DOI: 10.3390/medicina60020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Inflammatory bowel disease (IBD), especially Crohn's disease (CD), characterized by a chronic inflammatory process and progressive intestinal tissue damage, leads to the unrestrained proliferation of mesenchymal cells and the development of bowel strictures. Complications induced by fibrosis are related to high rates of morbidity and mortality and lead to a substantial number of hospitalizations and surgical procedures, generating high healthcare costs. The development of easily obtained, reliable fibrogenesis biomarkers is essential to provide an important complementary tool to existing diagnostic and prognostic methods in IBD management, guiding decisions on the intensification of pharmacotherapy, proceeding to surgical methods of treatment and monitoring the efficacy of anti-fibrotic therapy in the future. The most promising potential markers of fibrosis include cartilage oligomeric matrix protein (COMP), hepatocyte growth factor activator (HGFA), and fibronectin isoform- extra domain A (ED-A), as well as antibodies against granulocyte macrophage colony-stimulating factor (GM-CSF Ab), cathelicidin (LL-37), or circulatory miRNAs: miR-19a-3p and miR-19b-3p. This review summarizes the role of genetic predisposition, and risk factors and serological markers potentially contributing to the pathophysiology of fibrotic strictures in the course of IBD.
Collapse
|
35
|
Bai Z, Zhang D, Gao Y, Tao B, Bao S, Enninful A, Zhang D, Su G, Tian X, Zhang N, Xiao Y, Liu Y, Gerstein M, Li M, Xing Y, Lu J, Xu ML, Fan R. Spatially Exploring RNA Biology in Archival Formalin-Fixed Paraffin-Embedded Tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579143. [PMID: 38370833 PMCID: PMC10871202 DOI: 10.1101/2024.02.06.579143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Spatial transcriptomics has emerged as a powerful tool for dissecting spatial cellular heterogeneity but as of today is largely limited to gene expression analysis. Yet, the life of RNA molecules is multifaceted and dynamic, requiring spatial profiling of different RNA species throughout the life cycle to delve into the intricate RNA biology in complex tissues. Human disease-relevant tissues are commonly preserved as formalin-fixed and paraffin-embedded (FFPE) blocks, representing an important resource for human tissue specimens. The capability to spatially explore RNA biology in FFPE tissues holds transformative potential for human biology research and clinical histopathology. Here, we present Patho-DBiT combining in situ polyadenylation and deterministic barcoding for spatial full coverage transcriptome sequencing, tailored for probing the diverse landscape of RNA species even in clinically archived FFPE samples. It permits spatial co-profiling of gene expression and RNA processing, unveiling region-specific splicing isoforms, and high-sensitivity transcriptomic mapping of clinical tumor FFPE tissues stored for five years. Furthermore, genome-wide single nucleotide RNA variants can be captured to distinguish different malignant clones from non-malignant cells in human lymphomas. Patho-DBiT also maps microRNA-mRNA regulatory networks and RNA splicing dynamics, decoding their roles in spatial tumorigenesis trajectory. High resolution Patho-DBiT at the cellular level reveals a spatial neighborhood and traces the spatiotemporal kinetics driving tumor progression. Patho-DBiT stands poised as a valuable platform to unravel rich RNA biology in FFPE tissues to study human tissue biology and aid in clinical pathology evaluation.
Collapse
Affiliation(s)
- Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Dingyao Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yan Gao
- Center for Computational and Genomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bo Tao
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Shuozhen Bao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Archibald Enninful
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Daiwei Zhang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Graham Su
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Xiaolong Tian
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Ningning Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yang Liu
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mark Gerstein
- Section on Biomedical Informatics and Data Science, Yale University, New Haven, CT 06520, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Xing
- Center for Computational and Genomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Lu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mina L. Xu
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Human and Translational Immunology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
36
|
Hardisty G, Nicol MQ, Shaw DJ, Bennet ID, Bryson K, Ligertwood Y, Schwarze J, Beard PM, Hopkins J, Dutia BM. Latent gammaherpesvirus infection enhances type I IFN response and reduces virus spread in an influenza A virus co-infection model. J Gen Virol 2024; 105. [PMID: 38329395 DOI: 10.1099/jgv.0.001962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Infections with persistent or latent viruses alter host immune homeostasis and have potential to affect the outcome of concomitant acute viral infections such as influenza A virus (IAV). Gammaherpesviruses establish life-long infections and require an on-going immune response to control reactivation. We have used a murine model of co-infection to investigate the response to IAV infection in mice latently infected with the gammaherpesvirus MHV-68. Over the course of infection, latently infected BALB/c mice showed less weight loss, clinical signs, pulmonary cellular infiltration and expression of inflammatory mediators than naïve mice infected with IAV and had significantly more activated CD8+ T cells in the lungs. Four days after IAV infection, virus spread in the lungs of latently infected animals was significantly lower than in naïve animals. By 7 days after IAV infection latently infected lungs express elevated levels of cytokines and chemokines indicating they are primed to respond to the secondary infection. Investigation at an early time point showed that 24 h after IAV infection co-infected animals had higher expression of IFNβ and Ddx58 (RIG-I) and a range of ISGs than mice infected with IAV alone suggesting that the type I IFN response plays a role in the protective effect. This effect was mouse strain dependent and did not occur in 129/Sv/Ev mice. These results offer insight into innate immune mechanisms that could be utilized to protect against IAV infection and highlight on-going and persistent viral infections as a significant factor impacting the severity of acute respiratory infections.
Collapse
Affiliation(s)
- Gareth Hardisty
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 4-5 Little France Drive, Edinburgh. EH16 4UU, UK
| | - Marlynne Q Nicol
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Darren J Shaw
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Ian D Bennet
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Karen Bryson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Yvonne Ligertwood
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Jurgen Schwarze
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 4-5 Little France Drive, Edinburgh. EH16 4UU, UK
| | - Philippa M Beard
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
- School of Life Sciences, Keele University, Keele, Staffordshire, ST5 5BF, UK
| | - John Hopkins
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Bernadette M Dutia
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| |
Collapse
|
37
|
Amoriello R, Memo C, Ballerini L, Ballerini C. The brain cytokine orchestra in multiple sclerosis: from neuroinflammation to synaptopathology. Mol Brain 2024; 17:4. [PMID: 38263055 PMCID: PMC10807071 DOI: 10.1186/s13041-024-01077-7] [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: 11/21/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
The central nervous system (CNS) is finely protected by the blood-brain barrier (BBB). Immune soluble factors such as cytokines (CKs) are normally produced in the CNS, contributing to physiological immunosurveillance and homeostatic synaptic scaling. CKs are peptide, pleiotropic molecules involved in a broad range of cellular functions, with a pivotal role in resolving the inflammation and promoting tissue healing. However, pro-inflammatory CKs can exert a detrimental effect in pathological conditions, spreading the damage. In the inflamed CNS, CKs recruit immune cells, stimulate the local production of other inflammatory mediators, and promote synaptic dysfunction. Our understanding of neuroinflammation in humans owes much to the study of multiple sclerosis (MS), the most common autoimmune and demyelinating disease, in which autoreactive T cells migrate from the periphery to the CNS after the encounter with a still unknown antigen. CNS-infiltrating T cells produce pro-inflammatory CKs that aggravate local demyelination and neurodegeneration. This review aims to recapitulate the state of the art about CKs role in the healthy and inflamed CNS, with focus on recent advances bridging the study of adaptive immune system and neurophysiology.
Collapse
Affiliation(s)
- Roberta Amoriello
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy.
| | - Christian Memo
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy
| | - Laura Ballerini
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy
| | - Clara Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
| |
Collapse
|
38
|
Schwarz E, Benner B, Yu L, Tounkara F, Carson WE. Analysis of Changes in Plasma Cytokine Levels in Response to IL12 Therapy in Three Clinical Trials. CANCER RESEARCH COMMUNICATIONS 2024; 4:81-91. [PMID: 38108458 PMCID: PMC10777814 DOI: 10.1158/2767-9764.crc-23-0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 10/04/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The ability of IL12 to stimulate natural killer (NK) cell and T-cell antitumor activity makes it an attractive candidate for the immune therapy of cancer. Our group has demonstrated that IL12 enhances the NK cell response to antibody-coated tumor cells and conducted three clinical trials utilizing IL12 with mAbs (OSU-9968, OSU-0167, and OSU-11010). To better characterize IL12-induced immunity, plasma cytokine levels were measured in 21 patients from these trials with favorable and unfavorable responses. t-statistics and linear modeling were used to test for differences within and between response groups by examining levels at baseline and post-IL12 administration. Patients exhibited significant increases in 11 cytokines post-IL12 administration when analyzed collectively. However, several cytokines were differentially induced by IL12 depending on response. GMCSF was significantly increased in complete/partially responding patients, while stable disease patients had significant increases in IL10 and decreases in VEGF-C. Patients who experienced progressive disease had significant increases in CCL3, CCL4, IL18, TNFα, CXCL10, CCL8, CCL2, IL6, and IFNγ. The increases in CCL3, CCL4, and IL6 in progressive disease patients were significantly higher than in clinically benefitting patients and most prominent within the first two cycles of IL12 therapy. This correlative pilot study has identified changes that occur in levels of circulating cytokines following IL12 administration to patients with cancer, but this report must be viewed as exploratory in nature. It is meant to spark further inquiry into the topic via the analysis of additional cohorts of patients with similar characteristics who have received IL12 in a uniform fashion. SIGNIFICANCE IL12 activates immune cells and is used to treat cancer. The profile of circulating cytokines was measured in an exploratory fashion in patients with cancer that received IL12 in combination with mAbs. This correlative pilot study could serve as the basis for additional studies of IL12 effects on the production of immune cytokines.
Collapse
Affiliation(s)
- Emily Schwarz
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Brooke Benner
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Fode Tounkara
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - William E. Carson
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Department of Surgery, Division of Surgical Oncology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
39
|
Zhang H, Chen Y, Jiang X, Gu Q, Yao J, Wang X, Wu J. Unveiling the landscape of cytokine research in glioma immunotherapy: a scientometrics analysis. Front Pharmacol 2024; 14:1333124. [PMID: 38259287 PMCID: PMC10800575 DOI: 10.3389/fphar.2023.1333124] [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: 11/04/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Background: Cytokines modulate the glioma tumor microenvironment, influencing occurrence, progression, and treatment response. Strategic cytokine application may improve glioma immunotherapy outcomes. Gliomas remain refractory to standard therapeutic modalities, but immunotherapy shows promise given the integral immunomodulatory roles of cytokines. However, systematic evaluation of cytokine glioma immunotherapy research is absent. Bibliometric mapping of the research landscape, recognition of impactful contributions, and elucidation of evolutive trajectories and hot topics has yet to occur, potentially guiding future efforts. Here, we analyzed the structure, evolution, trends, and hotspots of the cytokine glioma immunotherapy research field, subsequently focusing on avenues for future investigation. Methods: This investigation conducted comprehensive bibliometric analyses on a corpus of 1529 English-language publications, from 1 January 2000, to 4 October 2023, extracted from the Web of Science database. The study employed tools including Microsoft Excel, Origin, VOSviewer, CiteSpace, and the Bibliometrix R package, to systematically assess trends in publication, contributions from various countries, institutions, authors, and journals, as well as to examine literature co-citation and keyword distributions within the domain of cytokines for glioma immunotherapy. The application of these methodologies facilitated a detailed exploration of the hotspots, the underlying knowledge structure, and the developments in the field of cytokines for glioma immunotherapy. Results: This bibliometric analysis revealed an exponential growth in annual publications, with the United States, China, and Germany as top contributors. Reviews constituted 17% and research articles 83% of total publications. Analysis of keywords like "interleukin-13," "TGF-beta," and "dendritic cells" indicated progression from foundational cytokine therapies to sophisticated understanding of the tumor microenvironment and immune dynamics. Key research avenues encompassed the tumor microenvironment, epidermal growth factor receptor, clinical trials, and interleukin pathways. This comprehensive quantitative mapping of the glioma immunotherapy cytokine literature provides valuable insights to advance future research and therapeutic development. Conclusion: This study has identified remaining knowledge gaps regarding the role of cytokines in glioma immunotherapy. Future research will likely focus on the tumor microenvironment, cancer vaccines, epidermal growth factor receptor, and interleukin-13 receptor alpha 2. Glioma immunotherapy development will continue through investigations into resistance mechanisms, microglia and macrophage biology, and interactions within the complex tumor microenvironment.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Chen
- Gamma Knife Center, Department of Oncology, Department of Neurological Surgery, Tianjin Huanhu Hospital, Tianjin Medical University, Tianjin, China
| | - Xinzhan Jiang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Qiang Gu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiahao Yao
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuefeng Wang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianghua Wu
- School of Nursing, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong, China
| |
Collapse
|
40
|
Chaker SC, Saad M, Mayes T, Lineaweaver WC. Burn Injury-related Growth Factor Expressions and Their Potential Roles in Burn-related Neuropathies. J Burn Care Res 2024; 45:25-31. [PMID: 37978864 DOI: 10.1093/jbcr/irad184] [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: 07/21/2023] [Indexed: 11/19/2023]
Abstract
In the context of burn injury, growth factors (GFs) play a significant role in mediating the complex local and systematic processes that occur. Among the many systemic complications that arise following a burn injury, peripheral neuropathy remains one of the most common. Despite the broad understanding of the effects GFs have on multiple tissues, their potential implications in both wound healing and neuropathy remain largely unexplored. Therefore, this review aims to investigate the expression patterns of GFs prominent during the burn wound healing process and explore the potential contributions these GFs have on the development of burn-related peripheral neuropathy.
Collapse
Affiliation(s)
- Sara C Chaker
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232USA
| | - Mariam Saad
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232USA
| | - Taylor Mayes
- Middle Tennessee State University, Murfreesboro, TN, 37132USA
| | - William C Lineaweaver
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232USA
| |
Collapse
|
41
|
White CS, Hung CC, Lanka S, Maddox CW, Barri A, Sokale AO, Dilger RN. Dietary monoglyceride supplementation to support intestinal integrity and host defenses in health-challenged weanling pigs. J Anim Sci 2024; 102:skae105. [PMID: 38629856 PMCID: PMC11044705 DOI: 10.1093/jas/skae105] [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: 02/02/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
Frequent incidence of postweaning enterotoxigenic Escherichia coli (ETEC) diarrhea in the swine industry contributes to high mortality rates and associated economic losses. In this study, a combination of butyric, caprylic, and capric fatty acid monoglycerides was investigated to promote intestinal integrity and host defenses in weanling pigs infected with ETEC. A total of 160 pigs were allotted to treatment groups based on weight and sex. Throughout the 17-d study, three treatment groups were maintained: sham-inoculated pigs fed a control diet (uninfected control [UC], n = 40), ETEC-inoculated pigs fed the same control diet (infected control [IC], n = 60), and ETEC-inoculated pigs fed the control diet supplemented with monoglycerides included at 0.3% of the diet (infected supplemented [MG], n = 60). After a 7-d acclimation period, pigs were orally inoculated on each of three consecutive days with either 3 mL of a sham-control (saline) or live ETEC culture (3 × 109 colony-forming units/mL). The first day of inoculations was designated as 0 d postinoculation (DPI), and all study outcomes reference this time point. Fecal, tissue, and blood samples were collected from 48 individual pigs (UC, n = 12; IC, n = 18; MG, n = 18) on 5 and 10 DPI for analysis of dry matter (DM), bacterial enumeration, inflammatory markers, and intestinal permeability. ETEC-inoculated pigs in both the IC and MG groups exhibited clear signs of infection including lower (P < 0.05) gain:feed and fecal DM, indicative of excess water in the feces, and elevated (P < 0.05) rectal temperatures, total bacteria, total E. coli, and total F18 ETEC during the peak-infection period (5 DPI). Reduced (P < 0.05) expression of the occludin, tumor necrosis factor α, and vascular endothelial growth factor A genes was observed in both ETEC-inoculated groups at the 5 DPI time point. There were no meaningful differences between treatments for any of the outcomes measured at 10 DPI. Overall, all significant changes were the result of the ETEC infection, not monoglyceride supplementation.
Collapse
Affiliation(s)
- Cameron S White
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Chien-Che Hung
- Veterinary Diagnostic Laboratory, University of Illinois, Urbana, IL, USA
| | - Saraswathi Lanka
- Veterinary Diagnostic Laboratory, University of Illinois, Urbana, IL, USA
| | - Carol W Maddox
- Veterinary Diagnostic Laboratory, University of Illinois, Urbana, IL, USA
| | | | | | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| |
Collapse
|
42
|
Almeida SFF, Santos L, Sampaio-Ribeiro G, Ferreira HRS, Lima N, Caetano R, Abreu M, Zuzarte M, Ribeiro AS, Paiva A, Martins-Marques T, Teixeira P, Almeida R, Casanova JM, Girão H, Abrunhosa AJ, Gomes CM. Unveiling the role of osteosarcoma-derived secretome in premetastatic lung remodelling. J Exp Clin Cancer Res 2023; 42:328. [PMID: 38031171 PMCID: PMC10688015 DOI: 10.1186/s13046-023-02886-9] [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] [Received: 05/18/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Lung metastasis is the most adverse clinical factor and remains the leading cause of osteosarcoma-related death. Deciphering the mechanisms driving metastatic spread is crucial for finding open therapeutic windows for successful organ-specific interventions that may halt or prevent lung metastasis. METHODS We employed a mouse premetastatic lung-based multi-omics integrative approach combined with clinical features to uncover the specific changes that precede lung metastasis formation and identify novel molecular targets and biomarker of clinical utility that enable the design of novel therapeutic strategies. RESULTS We found that osteosarcoma-bearing mice or those preconditioned with the osteosarcoma cell secretome harbour profound lung structural alterations with airway damage, inflammation, neutrophil infiltration, and extracellular matrix remodelling with increased deposition of fibronectin and collagens by resident stromal activated fibroblasts, favouring the adhesion of disseminated tumour cells. Systemic-induced microenvironmental changes, supported by transcriptomic and histological data, promoted and accelerated lung metastasis formation. Comparative proteome profiling of the cell secretome and mouse plasma identified a large number of proteins involved in extracellular-matrix organization, cell-matrix adhesion, neutrophil degranulation, and cytokine-mediated signalling, consistent with the observed lung microenvironmental changes. Moreover, we identified EFEMP1, an extracellular matrix glycoprotein exclusively secreted by metastatic cells, in the plasma of mice bearing a primary tumour and in biopsy specimens from osteosarcoma patients with poorer overall survival. Depletion of EFEMP1 from the secretome prevents the formation of lung metastasis. CONCLUSIONS Integration of our data uncovers neutrophil infiltration and the functional contribution of stromal-activated fibroblasts in ECM remodelling for tumour cell attachment as early pro-metastatic events, which may hold therapeutic potential in preventing or slowing the metastatic spread. Moreover, we identified EFEMP1, a secreted glycoprotein, as a metastatic driver and a potential candidate prognostic biomarker for lung metastasis in osteosarcoma patients. Osteosarcoma-derived secreted factors systemically reprogrammed the lung microenvironment and fostered a growth-permissive niche for incoming disseminated cells to survive and outgrow into overt metastasis. Daily administration of osteosarcoma cell secretome mimics the systemic release of tumour-secreted factors of a growing tumour in mice during PMN formation; Transcriptomic and histological analysis of premetastatic lungs revealed inflammatory-induced stromal fibroblast activation, neutrophil infiltration, and ECM remodelling as early onset pro-metastatic events; Proteome profiling identified EFEMP1, an extracellular secreted glycoprotein, as a potential predictive biomarker for lung metastasis and poor prognosis in osteosarcoma patients. Osteosarcoma patients with EFEMP1 expressing biopsies have a poorer overall survival.
Collapse
Affiliation(s)
- Sara F F Almeida
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, 3000-548, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
| | - Liliana Santos
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, 3000-548, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
| | - Gabriela Sampaio-Ribeiro
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
| | - Hugo R S Ferreira
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
| | - Nuno Lima
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
| | - Rui Caetano
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, 3004-561, Portugal
| | - Mónica Abreu
- Multidisciplinary Institute of Ageing (MIA), University of Coimbra, Coimbra, Portugal
| | - Mónica Zuzarte
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
| | - Ana Sofia Ribeiro
- Instituto de Investigação e Inovação em Saúde (i3S), Porto, 4200-135, Portugal
| | - Artur Paiva
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Tânia Martins-Marques
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
| | - Paulo Teixeira
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, 3004-561, Portugal
| | - Rui Almeida
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, 3004-561, Portugal
| | - José Manuel Casanova
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
- Tumor Unit of the Locomotor Apparatus (UTAL), Orthopedics Service, Coimbra Hospital and University Center (CHUC), University Clinic of Orthopedics, Coimbra, 3000-075, Portugal
| | - Henrique Girão
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal
| | - Antero J Abrunhosa
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, 3000-548, Portugal
| | - Célia M Gomes
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, 3000-548, Portugal.
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, 3000-548, Portugal.
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3000-075, Portugal.
| |
Collapse
|
43
|
Keturakis V, Narauskaitė D, Balion Z, Gečys D, Kulkovienė G, Kairytė M, Žukauskaitė I, Benetis R, Stankevičius E, Jekabsone A. The Effect of SARS-CoV-2 Spike Protein RBD-Epitope on Immunometabolic State and Functional Performance of Cultured Primary Cardiomyocytes Subjected to Hypoxia and Reoxygenation. Int J Mol Sci 2023; 24:16554. [PMID: 38068877 PMCID: PMC10705973 DOI: 10.3390/ijms242316554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/25/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Cardio complications such as arrhythmias and myocardial damage are common in COVID-19 patients. SARS-CoV-2 interacts with the cardiovascular system primarily via the ACE2 receptor. Cardiomyocyte damage in SARS-CoV-2 infection may stem from inflammation, hypoxia-reoxygenation injury, and direct toxicity; however, the precise mechanisms are unclear. In this study, we simulated hypoxia-reoxygenation conditions commonly seen in SARS-CoV-2-infected patients and studied the impact of the SARS-CoV-2 spike protein RBD-epitope on primary rat cardiomyocytes to gain insight into the potential mechanisms underlying COVID-19-related cardiac complications. Cell metabolic activity was evaluated with PrestoBlueTM. Gene expression of proinflammatory markers was measured by qRT-PCR and their secretion was quantified by Luminex assay. Cardiomyocyte contractility was analysed using the Myocyter plugin of ImageJ. Mitochondrial respiration was determined through Seahorse Mito Stress Test. In hypoxia-reoxygenation conditions, treatment of the SARS-CoV-2 spike RBD-epitope reduced the metabolic activity of primary cardiomyocytes, upregulated Il1β and Cxcl1 expression, and elevated GM-CSF and CCL2 cytokines secretion. Contraction time increased, while amplitude and beating frequency decreased. Acute treatment with a virus RBD-epitope inhibited mitochondrial respiration and lowered ATP production. Under ischaemia-reperfusion, the SARS-CoV-2 RBD-epitope induces cardiomyocyte injury linked to impaired mitochondrial activity.
Collapse
Affiliation(s)
- Vytenis Keturakis
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.K.)
- Department of Heart, Thoracic and Vascular Surgery, Medicine Faculty, Medical Academy, Lithuanian University of Health Sciences, 50103 Kaunas, Lithuania
| | - Deimantė Narauskaitė
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.K.)
| | - Zbigniev Balion
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.K.)
| | - Dovydas Gečys
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.K.)
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, 50103 Kaunas, Lithuania
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Lithuanian University of Health Sciences, 50166 Kaunas, Lithuania
| | - Gabrielė Kulkovienė
- Department of Drug Chemistry, Faculty of Pharmacy, Lithuanian University of Health Sciences, 50166 Kaunas, Lithuania
| | - Milda Kairytė
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.K.)
| | - Ineta Žukauskaitė
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.K.)
| | - Rimantas Benetis
- Department of Heart, Thoracic and Vascular Surgery, Medicine Faculty, Medical Academy, Lithuanian University of Health Sciences, 50103 Kaunas, Lithuania
| | - Edgaras Stankevičius
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.K.)
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Aistė Jekabsone
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.K.)
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Lithuanian University of Health Sciences, 50166 Kaunas, Lithuania
| |
Collapse
|
44
|
Sremac M, Luo H, Deng H, Parr MFE, Hutcheson J, Verde PS, Alagpulinsa DA, Kitzmann JM, Papas KK, Brauns T, Markmann JF, Lei J, Poznansky MC. Short-term function and immune-protection of microencapsulated adult porcine islets with alginate incorporating CXCL12 in healthy and diabetic non-human primates without systemic immune suppression: A pilot study. Xenotransplantation 2023; 30:e12826. [PMID: 37712342 DOI: 10.1111/xen.12826] [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: 04/20/2023] [Revised: 08/10/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023]
Abstract
Replacement of insulin-producing pancreatic beta-cells by islet transplantation offers a functional cure for type-1 diabetes (T1D). We recently demonstrated that a clinical grade alginate micro-encapsulant incorporating the immune-repellent chemokine and pro-survival factor CXCL12 could protect and sustain the integrity and function of autologous islets in healthy non-human primates (NHPs) without systemic immune suppression. In this pilot study, we examined the impact of the CXCL12 micro encapsulant on the function and inflammatory and immune responses of xenogeneic islets transplanted into the omental tissue bilayer sac (OB; n = 4) and diabetic (n = 1) NHPs. Changes in the expression of cytokines after implantation were limited to 2-6-fold changes in blood, most of which did not persist over the first 4 weeks after implantation. Flow cytometry of PBMCs following transplantation showed minimal changes in IFNγ or TNFα expression on xenoantigen-specific CD4+ or CD8+ T cells compared to unstimulated cells, and these occurred mainly in the first 4 weeks. Microbeads were readily retrievable for assessment at day 90 and day 180 and at retrieval were without microscopic signs of degradation or foreign body responses (FBR). In vitro and immunohistochemistry studies of explanted microbeads indicated the presence of functional xenogeneic islets at day 30 post transplantation in all biopsied NHPs. These results from a small pilot study revealed that CXCL12-microencapsulated xenogeneic islets abrogate inflammatory and adaptive immune responses to the xenograft. This work paves the way toward future larger scale studies of the transplantation of alginate microbeads with CXCL12 and porcine or human stem cell-derived beta cells or allogeneic islets into diabetic NHPs without systemic immunosuppression.
Collapse
Affiliation(s)
- Marinko Sremac
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hao Luo
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, China
| | - Hongping Deng
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Madeline F E Parr
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Pushkar S Verde
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David A Alagpulinsa
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jenna Miner Kitzmann
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, Arizona, USA
| | - Klearchos K Papas
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, Arizona, USA
| | - Timothy Brauns
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James F Markmann
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ji Lei
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [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: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
46
|
Okamoto M, Omori-Miyake M, Kuwahara M, Okabe M, Eguchi M, Yamashita M. The Inhibition of Glycolysis in T Cells by a Jak Inhibitor Ameliorates the Pathogenesis of Allergic Contact Dermatitis in Mice. J Invest Dermatol 2023; 143:1973-1982.e5. [PMID: 37028703 DOI: 10.1016/j.jid.2023.03.1667] [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: 09/13/2022] [Revised: 01/31/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023]
Abstract
Allergic contact dermatitis (ACD) and atopic dermatitis develop through delayed-type hypersensitivity reactions mediated by T cells. The development of immunomodulatory drugs, such as Jak inhibitors, would be useful for the long-term management of these diseases owing to their profile of favorable adverse effects. However, the efficacy of Jak inhibitors for ACD treatment has not been fully determined under a variety of settings. Therefore, we evaluated the effects of ruxolitinib, a Jak inhibitor for Jak1 and Jak2, using a mouse ACD model. As a result, the lower numbers of immune cells, including CD4+ T cells, CD8+ T cells, neutrophils, and possibly macrophages, as well as milder pathophysiological aspects have been observed in the inflamed skin of ACD with the administration of ruxolitinib. In addition, the treatment of differentiating T cells with ruxolitinib downregulated the level of IL-2-mediated glycolysis in vitro. Furthermore, symptoms of ACD did not develop in T-cell-specific Pgam1-deficient mice whose T cells had no glycolytic capacity. Taken together, our data suggest that the downregulation of glycolysis in T cells by ruxolitinib could be an important factor in the suppression of ACD development in mice.
Collapse
Affiliation(s)
- Michiko Okamoto
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Immunology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Miyuki Omori-Miyake
- Department of Infections and Host Defenses, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Makoto Kuwahara
- Department of Immunology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masataka Okabe
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Mariko Eguchi
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masakatsu Yamashita
- Department of Immunology, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Infections and Host Defenses, Ehime University Graduate School of Medicine, Ehime, Japan.
| |
Collapse
|
47
|
Sueblinvong V, Fan X, Hart C, Molina S, Koval M, Guidot DM. Ethanol-exposed lung fibroblasts cause airway epithelial barrier dysfunction. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1839-1849. [PMID: 37864530 DOI: 10.1111/acer.15174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Chronic alcohol ingestion predisposes to lung injury and disrepair during sepsis. Our previous studies outlined roles for transforming growth factor-beta 1 (TGFβ1) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in epithelial barrier homeostasis and how alcohol perturbs their expression and signaling. Here we hypothesize that ethanol-exposed lung fibroblasts (LF) are a source of dysregulated TGFβ1 and GM-CSF and thereby alter airway epithelial barrier function. METHODS Human or rat LF were cultured ± ethanol for 2 weeks and then co-cultured with human or rat airway epithelial cells (AEC) seeded on Transwell permeable supports. In selected groups, a TGFβ1 receptor type 1 (TGFβR1) inhibitor (SB431542) or a TGFβ1 neutralizing antibody was applied. Transepithelial electrical resistance (TER) was measured prior to co-culture and on day 5 of co-culture. AEC were then analyzed for the expression of selected tight junction and mesenchymal proteins, and transwell membranes were analyzed by immunofluorescence microscopy for ZO-1 expression and localization. TGFβ1 and GM-CSF levels in conditioned media from the co-cultures were quantified by ELISA. RESULTS AEC co-cultured with ethanol-exposed LF (ELF) showed a significant reduction in TER and corresponding decreases in ZO-1 expression, whereas collagen type 1A1 and α-smooth muscle actin protein expression were increased. In parallel, in conditioned media from the ELF + AEC co-cultures, activated TGFβ1 levels increased and GM-CSF levels decreased. Notably, all the effects of ELF on the AEC were prevented by blocking TGFβ1 activity. CONCLUSIONS Prior ethanol exposure to LF induces barrier dysfunction in naive AEC in a paracrine fashion through activation of TGFβ1 signaling and suppression of GM-CSF. These experimental findings provide a potential mechanism by which chronic alcohol ingestion impairs airway epithelial integrity and renders individuals susceptible to lung injury.
Collapse
Affiliation(s)
- Viranuj Sueblinvong
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Xian Fan
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Craishun Hart
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Samuel Molina
- FUJIFILM Irvine Scientific, Warminster, Pennsylvania, USA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David M Guidot
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
48
|
Min SJ, Kim SJ, Park JY, Seo CS, Choi YK. Preparation of Herbal Extracts for Intestinal Immune Modulation Activity Based on In Vitro Screening and In Vivo Evaluation of Zingiber officinale Rosc. Extracts. Molecules 2023; 28:6743. [PMID: 37764519 PMCID: PMC10536359 DOI: 10.3390/molecules28186743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Ten traditional herbal extracts effective against diarrhea, infectious diseases, and bacterial activity were selected and analyzed for Peyer's patch cell-mediated intestinal immunomodulatory activity in vitro and in vivo. Among the 10 herbal extracts, Zingiber officinale Rosc. (ZO) extract induced the highest secretion of immunoglobulin A (IgA) and granulocyte macrophage colony-stimulating factor (GM-CSF) in the cells of Peyer's patches. Furthermore, animal experiments showed that IA production was enhanced with the oral administration of ZO extract (100 mg/kg and 300 mg/kg) for 10 days. In addition, 6-, 8-, 10-gingerol, and 6-, 8-, 10-shogaol, the six major index compounds of ZO extract, were analyzed using HPLC. Our study findings confirm the intestinal immunomodulatory activity of ZO extract and lay a strong foundation for future analytical studies aimed at determining the active components of ZO extracts.
Collapse
Affiliation(s)
- Su Ji Min
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Sung Jin Kim
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Jun Yeon Park
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea
| | - Chang-Seob Seo
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - You-Kyong Choi
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
49
|
Dharra R, Kumar Sharma A, Datta S. Emerging aspects of cytokine storm in COVID-19: The role of proinflammatory cytokines and therapeutic prospects. Cytokine 2023; 169:156287. [PMID: 37402337 PMCID: PMC10291296 DOI: 10.1016/j.cyto.2023.156287] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/24/2023] [Indexed: 07/06/2023]
Abstract
COVID-19 has claimed millions of lives during the last 3 years since initial cases were reported in Wuhan, China, in 2019. Patients with COVID-19 suffer from severe pneumonia, high fever, acute respiratory distress syndrome (ARDS), and multiple-organ dysfunction, which may also result in fatality in extreme cases. Cytokine storm (CS) is hyperactivation of the immune system, wherein the dysregulated production of proinflammatory cytokines could result in excessive immune cell infiltrations in the pulmonary tissues, resulting in tissue damage. The immune cell infiltration could also occur in other tissues and organs and result in multiple organs' dysfunction. The key cytokines implicated in the onset of disease severity include TNF-α, IFN-γ, IL-6, IL-1β, GM-CSF, and G-CSF. Controlling the CS is critical in treating COVID-19 disease. Therefore, different strategies are employed to mitigate the effects of CS. These include using monoclonal antibodies directed against soluble cytokines or the cytokine receptors, combination therapies, mesenchymal stem cell therapy, therapeutic plasma exchange, and some non-conventional treatment methods to improve patient immunity. The current review describes the role/s of critical cytokines in COVID-19-mediated CS and the respective treatment modalities.
Collapse
Affiliation(s)
- Renu Dharra
- CSIR-Institute of Microbial Technology, Sector 39 A, Chandigarh 160036, India
| | - Anil Kumar Sharma
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Sonal Datta
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India.
| |
Collapse
|
50
|
Jha A, Larkin J, Moore E. SOCS1-KIR Peptide in PEGDA Hydrogels Reduces Pro-Inflammatory Macrophage Activation. Macromol Biosci 2023; 23:e2300237. [PMID: 37337867 DOI: 10.1002/mabi.202300237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Indexed: 06/21/2023]
Abstract
Macrophages modulate the wound healing cascade by adopting different phenotypes such as pro-inflammatory (M1) or pro-wound healing (M2). To reduce M1 activation, the JAK/STAT pathway can be targeted by using suppressors of cytokine signaling (SOCS1) proteins. Recently a peptide mimicking the kinase inhibitory region (KIR) of SOCS1 has been utilized to manipulate the adaptive immune response. However, the utilization of SOCS1-KIR to reduce pro-inflammatory phenotype in macrophages is yet to be investigated in a biomaterial formulation. This study introduces a PEGDA hydrogel platform to investigate SOCS1-KIR as a macrophage phenotype manipulating peptide. Immunocytochemistry, cytokine secretion assays, and gene expression analysis for pro-inflammatory macrophage markers in 2D and 3D experiments demonstrate a reduction in M1 activation due to SOCS1-KIR treatment. The retention of SOCS1-KIR in the hydrogel through release assays and diffusion tests is demonstrated. The swelling ratio of the hydrogel also remains unaffected with the entrapment of SOCS1-KIR. This study elucidates how SOCS1-KIR peptide in PEGDA hydrogels can be utilized as an effective therapeutic for macrophage manipulation.
Collapse
Affiliation(s)
- Aakanksha Jha
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Joseph Larkin
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, 32603, USA
| | - Erika Moore
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, 20742, USA
| |
Collapse
|