1
|
Foulem RD, Mbarik M, Doiron JA, Soucy MFN, Toro-Ramirez D, Pecourt F, Barnett DA, Boudreau LH, Surette ME. Platelet-derived microvesicles modulate cytokine and lipid mediator profiles in THP-1 monocytes and macrophages. Immunol Lett 2025; 275:107029. [PMID: 40306329 DOI: 10.1016/j.imlet.2025.107029] [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: 12/17/2024] [Revised: 04/07/2025] [Accepted: 04/26/2025] [Indexed: 05/02/2025]
Abstract
Monocytes are circulating immune cells that migrate to inflamed tissues and differentiate into macrophages, where they play a dual role in regulating pro-inflammatory and pro-resolving responses through cytokine and lipid mediator secretion. Platelet-derived microvesicles (PMVs), released during platelet activation, infiltrate inflamed areas and interact with monocytes and macrophages, facilitating the transfer of bioactive contents. While these interactions have been observed, their functional consequences on monocyte/macrophage inflammatory profiles remain poorly understood. In this study, PMVs are shown to be internalized by human THP-1 monocytes. The interaction with THP-1 cells occurs rapidly, with 60 % of cells interacting with PMVs within one hour. When cells are differentiated to M0 and M1 macrophages, interactions with PMVs only peak after 24 h. Interaction of cells with PMVs resulted in an increased capacity to synthesize cyclooxygenase- and lipoxygenase-derived lipid mediators of inflammation, especially in M1 cells. Cytokine production was also influenced in a cell-state-dependent manner. PMVs had no impact on undifferentiated THP-1 cells but enhanced the production of several cytokines in M0 cells as well as IL-23 and IL-6 in M1 macrophages. When stimulated with lipopolysaccharides, PMV-treated M0 macrophages demonstrated elevated production of the anti-inflammatory cytokine IL-10, while M1 macrophages exhibited increased secretion of IL-1β, MCP-1, and IL-6, highlighting an effect on pro-inflammatory cytokine production. These findings reveal that PMVs selectively modulate the inflammatory cytokine and lipid mediator profiles of monocytes and macrophages depending on their differentiation state. This study underscores the role of PMVs as key players in intercellular communication and immune regulation, particularly in the context of inflammation.
Collapse
Affiliation(s)
- Robert D Foulem
- Department of Chemistry and Biochemistry, Université de Moncton, Canada; New Brunswick Center for Precision Medicine, Moncton, Canada
| | - Maroua Mbarik
- Department of Chemistry and Biochemistry, Université de Moncton, Canada; New Brunswick Center for Precision Medicine, Moncton, Canada
| | - Jérémie A Doiron
- Department of Chemistry and Biochemistry, Université de Moncton, Canada; New Brunswick Center for Precision Medicine, Moncton, Canada; Atlantic Cancer Research Institute, Moncton, Canada
| | - Marie-France N Soucy
- Department of Chemistry and Biochemistry, Université de Moncton, Canada; New Brunswick Center for Precision Medicine, Moncton, Canada
| | - Dayana Toro-Ramirez
- Department of Chemistry and Biochemistry, Université de Moncton, Canada; New Brunswick Center for Precision Medicine, Moncton, Canada; Universidad de Antioquia, Medellín, Colombia
| | - Florient Pecourt
- Department of Chemistry and Biochemistry, Université de Moncton, Canada; New Brunswick Center for Precision Medicine, Moncton, Canada; Aix-Marseille Université, Marseille, France
| | | | - Luc H Boudreau
- Department of Chemistry and Biochemistry, Université de Moncton, Canada; New Brunswick Center for Precision Medicine, Moncton, Canada
| | - Marc E Surette
- Department of Chemistry and Biochemistry, Université de Moncton, Canada; New Brunswick Center for Precision Medicine, Moncton, Canada.
| |
Collapse
|
2
|
Wang H, Wang S, Wang J, Fang Y, Li J, Shen Y, Guo J. Sorafenib inhibits multiple sclerosis by regulating T cell differentiation. Cell Signal 2025; 133:111872. [PMID: 40389044 DOI: 10.1016/j.cellsig.2025.111872] [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: 01/11/2025] [Revised: 04/23/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
Multiple sclerosis (MS) is a group of disorder characterized by aberrant T cell reactivity toward self-antigens with loss of immunological tolerance, resulting in chronic inflammation and tissue damage. CD4+ Th cells can differentiate into Th1, Th2, Th17, and Treg cells in response to a specific class of pathogenic microorganisms and to the cytokine milieu. Here, we found that tyrosine kinase inhibitor sorafenib (Sora), which had been approved by FDA for the treatment of tumor, could suppress pro-inflammatory Th1, Th17 cell differentiation, and promote anti-inflammatory Treg cell polarization. Furthermore, Sora suppressed Th1 and Th17 cell differentiation by STAT4 and TGF-β1 signaling, respectively. In addition, treatment with Sora in mice inhibited Th1, Th17 cell accumulation and promoted Treg cell gather in the brain, thus protecting mice from experimental autoimmune encephalomyelitis (EAE). These results suggest that Sora may be a potential treatment for autoimmune diseases.
Collapse
MESH Headings
- Animals
- Sorafenib/pharmacology
- Cell Differentiation/drug effects
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Mice
- Th17 Cells/drug effects
- Th17 Cells/cytology
- Th17 Cells/immunology
- Mice, Inbred C57BL
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/cytology
- Th1 Cells/drug effects
- Th1 Cells/cytology
- Th1 Cells/immunology
- Signal Transduction/drug effects
- Transforming Growth Factor beta1/metabolism
- Female
- STAT4 Transcription Factor/metabolism
- Phenylurea Compounds/pharmacology
- Niacinamide/analogs & derivatives
- Niacinamide/pharmacology
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Hanliang Wang
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China; Department of Medical Oncology, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, Zhejiang 310020, China
| | - Shuowang Wang
- Medical School of Zhejiang University, Hangzhou, Zhejiang 310020, China
| | - Jin Wang
- Department of Medical Oncology, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, Zhejiang 310020, China
| | - Yue Fang
- Medical School of Zhejiang University, Hangzhou, Zhejiang 310020, China
| | - Junwei Li
- Medical School of Zhejiang University, Hangzhou, Zhejiang 310020, China
| | - Yingying Shen
- Department of Medical Oncology, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, Zhejiang 310020, China.
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
3
|
Vinh DC. Human immunity to fungal infections. J Exp Med 2025; 222:e20241215. [PMID: 40232283 PMCID: PMC11998751 DOI: 10.1084/jem.20241215] [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: 12/02/2024] [Revised: 01/07/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Fungi increasingly threaten health globally. Mycoses range from life-threatening, often iatrogenic conditions, to enigmatic syndromes occurring without apparent immunosuppression. Despite some recent advances in antifungal drug development, complementary therapeutic strategies are essential for addressing these opportunistic pathogens. One promising avenue is leveraging host immunity to combat fungal infections; this necessitates deeper understanding of the molecular immunology of human fungal susceptibility to differentiate beneficial versus harmful immunopathological responses. Investigating human models of fungal diseases in natural settings, particularly through genetic immunodeficiencies and ethnographic-specific genetic vulnerabilities, reveals crucial immune pathways essential for fighting various yeasts and molds. This review highlights the diversity in intrinsic fungal susceptibility across individuals and populations, through genetic- and autoantibody-mediated processes, complementing previous principles learned from animal studies and iatrogenic contexts. Improved understanding of human immunity to fungal diseases will facilitate the development of host-directed immunotherapies and targeted public health interventions, paving the way for precision medicine in fungal disease management.
Collapse
Affiliation(s)
- Donald C. Vinh
- Department of Medicine (Division of Infectious Diseases), McGill University Health Center, Montreal, Canada
- Department of OptiLab (Division of Medical Microbiology, Division of Molecular Genetics-Immunology), McGill University Health Center, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
- Center of Reference for Genetic Research in Infection and Immunity, McGill University Health Center Research Institute, Montreal, Canada
| |
Collapse
|
4
|
Becker SL, Vague M, Ortega-Loayza AG. Insights into the Pathogenesis of Pyoderma Gangrenosum. J Invest Dermatol 2025; 145:1305-1322. [PMID: 39718519 DOI: 10.1016/j.jid.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/31/2024] [Accepted: 09/25/2024] [Indexed: 12/25/2024]
Abstract
Pyoderma gangrenosum (PG) is a neutrophilic dermatosis of unclear etiology. Numerous theories of its underlying pathogenesis have been proposed, including external triggers, neutrophilic dysfunction, complement activation, and autoimmunity, as well as a possible component of underlying genetic susceptibility. This review seeks to synthesize current understanding of the pathogenesis of PG and integrate interactions between the multitude of implicated host immune pathways to guide and inform future directions into the treatment of PG.
Collapse
Affiliation(s)
- Sarah L Becker
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Morgan Vague
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Alex G Ortega-Loayza
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA.
| |
Collapse
|
5
|
Iborra‐Palau EV, Garcia‐Redondo E, Alabau‐Dasi R. Factors Influencing Adherence to Phototherapy in Patients With Psoriasis: A Cross-Sectional Study. J Adv Nurs 2025; 81:3110-3117. [PMID: 39315759 PMCID: PMC12080098 DOI: 10.1111/jan.16472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024]
Abstract
AIMS Assess the level of adherence to phototherapy and determine what factors influence it. DESIGN Cross-sectional. METHODS This study included a convenience sampling of 72 patients with psoriasis undergoing phototherapy. Data were collected through a self-reported questionnaire with sociodemographic variables, the Goldberg Anxiety and Depression Scale, the Short Form Health Survey and the Dermatology Life Quality Index. Adherence to the treatment and its ending was measured through a session record. RESULTS A small percentage of the participants demonstrated adequate adherence, and nearly half of them had low adherence. The factors statistically significant and with a negative impact on adherence were as follows: having a partner, experiencing anxiety or depression or using public transportation to get to the hospital. The probability of not adhering to the treatment increased when: patients found it difficult to attend therapy; perceiving their mental and physical health as being worse; experiencing anxiety or depression; having a diagnosed mental illness; being a man; or having had the sickness for an extended period of time. CONCLUSION This study determined the level of adherence to phototherapy and advanced our understanding of this variable. Women exhibited higher levels of adherence compared to men, although they reported worse perceived mental and physical health, and the disease had a higher impact on their life. IMPLICATIONS FOR THE PROFESSION AND/OR PATIENT CARE Informing phototherapy nurses on the factors that impact treatment adherence may help to increase the treatment compliance, which may improve psoriasis patients' clinical symptoms. IMPACT Increase the body of knowledge about the treatment that phototherapy nurses administer. REPORTING METHOD STORBE guidelines. PATIENT OR PUBLIC CONTRIBUTION No patient or public contribution.
Collapse
Affiliation(s)
- Elena Violeta Iborra‐Palau
- Department of Nursing, School of Nursing and PodiatryUniversity of ValenciaValenciaSpain
- General University Hospital of ValenciaValenciaSpain
| | - Elena Garcia‐Redondo
- Department of Nursing, School of Nursing and PodiatryUniversity of ValenciaValenciaSpain
- Malvarrosa HospitalValenciaSpain
| | - Raquel Alabau‐Dasi
- Department of Nursing and Podiatry, School of Health SciencesUniversity of MalagaMalagaSpain
| |
Collapse
|
6
|
Hou J, Zhao J, He H, Ma W, Wang D, Peng X, Ge X, Chen J. Distribution of two CD4 +FOXP3 + T cell subpopulations reflects disease phenotypes and prognosis in COPD. Sci Rep 2025; 15:17721. [PMID: 40399434 PMCID: PMC12095526 DOI: 10.1038/s41598-025-02935-7] [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: 11/14/2024] [Accepted: 05/16/2025] [Indexed: 05/23/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous condition, with varying clinical phenotypes and prognoses. Regulatory T cells (Tregs), particularly CD4+FOXP3+ T cell subpopulations, are crucial in modulating immune responses. This study investigates the distribution of two CD4+FOXP3+ T cell subpopulations in bronchoalveolar lavage fluid (BALF) from COPD patients and their association with disease phenotypes and prognosis. Patients were classified into Type A (lower frequencies of inflammatory FOXP3lo T cells) and Type B (higher frequencies of inflammatory FOXP3lo T cells). Type B COPD patients, who demonstrated more severe emphysema, heightened inflammatory responses, faster lung function decline, and more pronounced osteoporosis, showed a significant increase in FOXP3lo non-suppressive T cells. In contrast, Type A patients exhibited a higher proportion of FOXP3hi Treg cells, which correlated with milder disease phenotypes. The distinct distribution of CD4+FOXP3+ T cell subpopulations provides insights into the progression of COPD and suggests that these cells could serve as potential biomarkers for disease severity and prognosis. Further research may offer new therapeutic avenues by targeting these Treg subpopulations in COPD management.
Collapse
Affiliation(s)
- Jia Hou
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia, China.
| | - Jiale Zhao
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia, China
| | - Hua He
- Department of Infectious Disease, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang City, China
| | - Weirong Ma
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia, China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia, China
| | - Xinru Peng
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia, China
| | - Xiahui Ge
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai, China.
| | - Juan Chen
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia, China
- Department of Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
7
|
Karpoff K, Sun L, Cain CL, Valentine B, Simpson A, Ortega-Loayza AG. Paws to Patients: Canine Pyoderma Gangrenosum as a Reflective Model of Human Pyoderma Gangrenosum. Int J Dermatol 2025. [PMID: 40400070 DOI: 10.1111/ijd.17863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/23/2025]
Abstract
Human pyoderma gangrenosum (hPG) is a rare, non-infectious neutrophilic dermatosis characterized by painful, necrotic ulcers with violaceous borders. Research on hPG is limited by minimal existing animal models, the sole being a pharmacologically induced murine model. Few cases of canine pyoderma gangrenosum (cPG) have been published, and dogs are genetically closer to humans than are mice, making them a potentially valuable model of hPG. We aimed to characterize all published cPG cases and compare their presentation to that of hPG. A Google Scholar, PubMed, and Embase search of "canine AND pyoderma gangrenosum" was conducted in August 2024, with seven case reports, two case series, and two veterinarian-sourced cases ultimately reviewed. Of the 31 analyzed cPG cases, 30 presented with skin ulcers at multiple sites, the most common being the trunk (64.5%), paws (41.9%), and limbs (32.3%), mirroring the typical localization of hPG on the lower extremities, back, and abdomen. The average age of cPG presentation was 7.8 +/- 3.7 years (range: 0.25-14.5 years). Dogs reflected hPG comorbidities, with 16.1% presenting with gastrointestinal pathologies, 6.5% with arthritis, and 6.5% with hematologic disorders. Dogs were responsive to medications frequently employed in hPG-prednisone/prednisolone (80.6%) as initial therapy, and cyclosporine (77.4%) as maintenance therapy. Overall, cPG resembles hPG in affected anatomic sites, comorbidities, and therapeutic response. It offers a representative, spontaneously occurring hPG animal model alternative to the existing murine model. Future studies are necessary to reproduce cPG to further investigate disease pathophysiology.
Collapse
Affiliation(s)
- Kateryna Karpoff
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, New York, USA
| | - Lori Sun
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Christine L Cain
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Becky Valentine
- PETDERM, Calgary Pet Dermatology Centre, Calgary, Alberta, Canada
| | - Andrew Simpson
- Department of Dermatology, VCA Aurora Animal Hospital, Aurora, Illinois, USA
| | - Alex G Ortega-Loayza
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
8
|
Teuscher JL, Lupatsii M, Graspeuntner S, Jonassen S, Bringewatt A, Herting E, Stichtenoth G, Bossung V, Rupp J, Härtel C, Demmert M. Persistent reduction of Bifidobacterium longum in the infant gut microbiome in the first year of age following intrapartum penicillin prophylaxis for maternal GBS colonization. Front Immunol 2025; 16:1540979. [PMID: 40443663 PMCID: PMC12119681 DOI: 10.3389/fimmu.2025.1540979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Group B Streptococcus is a significant cause of early-onset disease in term newborns, with a global incidence of 0.41/1000 live births. Intrapartum antibiotic prophylaxis (IAP) has reduced EOD incidence by over 80%, but concerns exist about its impact on the neonatal gut microbiome and potential long-term health effects. Methods This single center study examines the effects of IAP on the fecal infant microbiome in the first year of age and on the T cell phenotype in the first days after birth among 22 infants receiving IAP with penicillin due to maternal GBS colonization and 26 infants not exposed to IAP. The fecal microbiome was analyzed at birth, one month and one year of age through 16S rRNA gene sequencing. Additionally, a T cell phenotyping of peripheral blood was performed between the second and fifth day of age. Results At one month, IAP exposed infants had a significantly lower relative abundance of Bifidobacterium longum in fecal samples, an effect which was sustained at one year. In IAP exposed infants we found a proinflammatory T-helper cell profile, characterized by higher IL-17A, RORgt, and TGF-b expression. Discussion This study proposes a sustained impact of IAP on the neonatal microbiome and T cell repertoire.
Collapse
Affiliation(s)
- Jana Lucia Teuscher
- Clinic for Pediatric and Adolescent Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Mariia Lupatsii
- Department for Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Simon Graspeuntner
- Department for Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Lübeck, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
- Medical Clinic III, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Sinje Jonassen
- Clinic for Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Arne Bringewatt
- Clinic for Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Egbert Herting
- Clinic for Pediatric and Adolescent Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Guido Stichtenoth
- Clinic for Pediatric and Adolescent Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Verena Bossung
- Clinic for Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Jan Rupp
- Department for Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Lübeck, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Christoph Härtel
- Pediatric Clinic and Policlinic, University Hospital Würzburg, Würzburg, Germany
| | - Martin Demmert
- Clinic for Pediatric and Adolescent Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
9
|
Oshika, Bari VK. Molecular mechanism of host-yeast interactions and prevention by nanoformulation approaches. Microb Pathog 2025; 205:107663. [PMID: 40339625 DOI: 10.1016/j.micpath.2025.107663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
Fungal infections are a major source of morbidity and mortality in people with compromised immune systems, such as those with human immunodeficiency virus, cancer, organ transplant recipients, and patients undergoing chemotherapy in healthcare settings. According to a recent World Health Organization (WHO) fungal priority pathogens list, Cryptococcus spp., Candida spp., Aspergillus spp., and Candida auris cause severe invasive infections in human. These opportunistic pathogens cause a significant number of mycoses, which affect over a billion people annually. Around two million infections can be fatal, especially for those with compromised immune systems. To diagnose and treat mycoses, we need to understand the complex interactions between the fungus and the host during pathogenesis, the virulence-causing traits of the fungus, and how the host fights infection through the immune system. Although several antifungal drugs are available against fungal infections, their effectiveness is highly variable, with adverse effects. In addition, the increasing resistance to traditional antifungal treatments poses serious risks to the healthcare industry. Therefore, new therapeutic strategies are required to combat these potentially fatal fungal infections. Nanostructure-based formulations can improve the therapeutic efficacy of conventional medications by broadening their activities, decreasing toxicity, enhancing bioactivity, and improving biodistribution. The review highlights host and fungus interaction and how nanoformulations can be targeted against fungal infections.
Collapse
Affiliation(s)
- Oshika
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO, Ghudda, Bathinda, India
| | - Vinay Kumar Bari
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO, Ghudda, Bathinda, India.
| |
Collapse
|
10
|
van der Werf J, Fleming NI. Are single nucleotide polymorphisms underutilized for guiding treatment of inflammatory bowel disease? Immunol Cell Biol 2025. [PMID: 40313162 DOI: 10.1111/imcb.70029] [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: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD), ulcerative colitis (UC) and IBD unclassified (IBDU), significantly impacts quality of life. Despite significant advances in the management of the conditions, responses to treatments vary greatly, and this is due partly to our natural genetic variation. Here we will review the evidence for whether single nucleotide polymorphisms (SNPs) have the potential to guide treatment decisions for people with IBD. We will first consider SNPs that exhibit strong associations with IBD pathogenesis and their relevance to epithelial barrier integrity, cytokine production, and immune system function. Then, we will cover those SNPs implicated in altering response to our various current IBD therapeutics, including the recently implemented drugs ustekinumab and tofacitinib. Finally, we will explore lesser-known SNPs that exhibit complex relationships with the disease and which may be undervalued as pharmacogenetic tools. Overall, it will be demonstrated that SNPs associated with IBD pathology are largely distinct from those predicting response to treatments and that new discoveries of clinically useful tools can be expected from therapy-focused investigations. Given the growing list of treatments available, we argue that beneficial personalization of treatments based on SNPs is still underutilized.
Collapse
Affiliation(s)
| | - Nicholas Ian Fleming
- Department of Pathology, University of Otago, Dunedin, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
11
|
Gossec L, Baraliakos X, Aletaha D, Sharaf M, Rampakakis E, Lavie F, López-Medina C, Selmi C, Coates LC. Multi-domain effectiveness of guselkumab evaluated via composite indices through 1 year in patients with PsA and inadequate response to TNFi: post hoc analysis of COSMOS. Rheumatology (Oxford) 2025; 64:2565-2574. [PMID: 39437003 PMCID: PMC12048074 DOI: 10.1093/rheumatology/keae586] [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: 08/22/2024] [Revised: 09/30/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
OBJECTIVE Evaluate guselkumab efficacy, an anti-interleukin-23p19-subunit antibody, in patients with active psoriatic arthritis (PsA) and inadequate response to 1 or 2 tumour necrosis factor inhibitors (TNFi-IR), utilizing composite indices assessing disease activity across disease domains. METHODS In the Phase IIIb COSMOS trial, 285 adults with TNFi-IR PsA were randomized (2:1) to receive guselkumab 100 mg or placebo at Week (W)0, W4, then every 8 weeks through W44. Patients receiving placebo crossed over to guselkumab at W24. In this post hoc analysis, composite indices evaluated included the Disease Activity Index for Psoriatic Arthritis (DAPSA), Disease Activity Score 28 (DAS28), Psoriatic Arthritis Response Criteria (PsARC), Psoriatic Arthritis Disease Activity Score (PASDAS), GRAPPA Composite score (GRACE), modified Composite Psoriatic Disease Activity Index (mCPDAI), minimal disease activity (MDA), and very low disease activity (VLDA). Through W24, treatment failure rules were applied. Through W48, non-responder imputation was used for missing data. RESULTS Greater proportions of guselkumab- than placebo-randomized patients achieved composite index endpoints relating to low disease activity (LDA; 14.8-52.4% vs 3.1-28.1%) or remission (3.7-5.3% vs 0.0-2.1%) at W24. Among guselkumab-randomized patients, LDA rates increased to W48 (DAPSA, 44.4%; DAS28, 47.8%; PASDAS, 34.4%; GRACE, 33.3%; mCPDAI, 40.2%), and 27.0% and 64.0% achieved MDA and a PsARC response, respectively. In the placebo→guselkumab crossover group, W48 response rates were similar to the guselkumab-randomized group. CONCLUSION Guselkumab treatment provided substantial benefits across multiple disease domains, with increasing proportions of patients achieving LDA/remission over 1 year, highlighting the effectiveness of guselkumab despite previous inadequate response to TNFi.
Collapse
Affiliation(s)
- Laure Gossec
- INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique, Sorbonne Universite, Paris, France
- APHP, Rheumatology Department, Pitie Salpetriere Hospital, Paris, France
| | | | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Emmanouil Rampakakis
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- JSS Medical Research Inc., Scientific Affairs, Montreal, QC, Canada
| | - Frédéric Lavie
- The Janssen Pharmaceutical Companies of Johnson & Johnson, Paris, France
| | | | - Carlo Selmi
- Rheumatology and Clinical Immunology, Humanitas Research Hospital, Rozzano, Milan, Italy
- Rheumatology and Clinical Immunology, Humanitas University, Milan, Italy
| | - Laura C Coates
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
12
|
Paton H, Sarkar P, Gurung P. An overview of host immune responses against Leishmania spp. infections. Hum Mol Genet 2025:ddaf043. [PMID: 40287829 DOI: 10.1093/hmg/ddaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Leishmania spp. infections pose a significant global health challenge, affecting approximately 1 billion people across more than 88 endemic countries. This unicellular, obligate intracellular parasite causes a spectrum of diseases, ranging from localized cutaneous lesions to systemic visceral infections. Despite advancements in modern medicine and increased understanding of the parasite's etiology and associated diseases, treatment options remain limited to pentavalent antimonials, liposomal amphotericin B, and miltefosine. A deeper understanding of the interactions between immune and non-immune cells involved in the clearance of Leishmania spp. infections could uncover novel therapeutic strategies for this debilitating disease. This review highlights recent progress in elucidating how various cell types contribute to the regulation and resolution of Leishmania spp. infections.
Collapse
Affiliation(s)
- Hanna Paton
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
| | - Prabuddha Sarkar
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Center for Immunology and Immune Based Disease, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Iowa City Veterans Affairs (VA) Medical Center, 601 US-6, Iowa City, IA 52246, United States
| |
Collapse
|
13
|
Koul R, Lu W, Torres T, Choi YB. Temporal relationship between neurofilament light chain and cytokines involved in T helper-17 lymphocyte signaling in the blood of experimental autoimmune encephalomyelitis mice. Mult Scler Relat Disord 2025; 99:106463. [PMID: 40279778 DOI: 10.1016/j.msard.2025.106463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/25/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune neuroinflammatory disease leading to eventual neurodegeneration. There is a strong interest in using blood biomarkers to detect the disease onset and monitor disease progression in MS. One such biomarker is neurofilament light chain (NfL) that reflects axonal damage or neurodegeneration. Proinflammatory cytokines involved in T-helper 17 (Th17) lymphocyte signaling are potential blood biomarkers of neuroinflammation. METHODS We induced experimental autoimmune encephalomyelitis (EAE), a mouse model for MS, in female C57BL/6 mice and measured the progression of EAE using a clinical score. We collected blood from EAE mice on the post-immunization days 7, 10, 16, or 22 and measured NfL concentrations using single molecule array (SIMOA) technology. We also measured concentrations of cytokines related to Th17 using xMAP immune assay technology (Luminex) from the same mice. RESULTS We found that the concentration of NfL began to rise at the onset of clinical symptoms on the post-immunization day 10 and peaked on day 16, closely correlating with the clinical score. However, the concentrations of several proinflammatory cytokines, including IFN-γ, IL-6, IL-17A, IL-22, and TNF-α, were already elevated even before the onset of the clinical symptoms and did not correlate positively with the clinical scores. CONCLUSION Our results confirmed the utility of NfL as a blood biomarker for disease progression in MS. The elevation of multiple proinflammatory cytokines concentrations in the blood of the same EAE mice in the presymptomatic stage suggested that blood biomarkers for neuroinflammation increased before blood biomarkers for neurodegeneration during the disease course in EAE.
Collapse
Affiliation(s)
- Raveesh Koul
- Neurology Service, VA New Jersey Health Care System, 385 Tremont Ave. East Orange, NJ 07018, USA
| | - Wei Lu
- Neurology Service, VA New Jersey Health Care System, 385 Tremont Ave. East Orange, NJ 07018, USA
| | - Timothy Torres
- Neurology Service, VA New Jersey Health Care System, 385 Tremont Ave. East Orange, NJ 07018, USA
| | - Yun-Beom Choi
- Neurology Service, VA New Jersey Health Care System, 385 Tremont Ave. East Orange, NJ 07018, USA; Department of Neurology, Rutgers New Jersey Medical School, 185 S. Orange Ave. Newark, NJ 07103, USA.
| |
Collapse
|
14
|
Gatica S, Paillal N, Rangel-Ramírez MA, Méndez L, Fernández-Tello A, Kalergis AM, Bueno SM, González PA, Soto JA, Simon F, Carreño LJ, Melo-Gonzalez F, Riedel CA. Gestational Hypothyroxinemia Shifts Th1/Th17 Immunity and Innate Lymphoid Cell Balance in the Adult Offspring during the Presymptomatic Stage of Experimental Autoimmune Encephalomyelitis. Neuroimmunomodulation 2025; 32:126-138. [PMID: 40209697 DOI: 10.1159/000545578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
INTRODUCTION Thyroid hormone homeostasis during pregnancy is crucial for proper neurodevelopment and cognitive capacity during adulthood. Accumulating evidence reveals that gestational hypothyroxinemia (HTX) modulates the immune response of the adult offspring. METHODS In the present study, adult mice gestated in HTX and their euthyroid counterparts were induced with a mild form of experimental autoimmune encephalomyelitis (EAE), a widespread model of multiple sclerosis, and analyzed at baseline and 7 days after EAE induction. RESULTS Levels of circulating IL-17 were significantly lower in mice gestated in HTX at both timepoints, while circulating IFN-γ was significantly higher only in mice gestated in HTX, 7 days after EAE induction. A significant increase in type 1 innate lymphoid cells (ILC1) was found only in mice gestated in HTX 7 days after EAE induction, while type 3 innate lymphoid cells (ILC3) populations showed no variation. Interestingly, a significant increase of Th17 CD4+ cells was found only in mice of euthyroid gestation, 7 days after EAE induction. CONCLUSION These results highlight the repercussions of thyroid hormone impairment in utero at adult ages while dissecting on the pathogenesis of EAE in terms of Th1/Th17 balance from an innate immune perspective. These findings contribute to the advancement of our comprehension of the presymptomatic stage of EAE, unveiling new paths for basic and translational research in the field of neuroinflammation.
Collapse
Affiliation(s)
- Sebastian Gatica
- Laboratorio de Endocrino-Inmunología, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Nicolas Paillal
- Laboratorio de Endocrino-Inmunología, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Ma Andreina Rangel-Ramírez
- Laboratorio de Endocrino-Inmunología, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Luis Méndez
- Laboratorio de Endocrino-Inmunología, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Odontología, Laboratorio de Odontología Traslacional, Universidad Andres Bello, Santiago, Chile
| | - Alonso Fernández-Tello
- Laboratorio de Endocrino-Inmunología, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Translational Immunology Laboratory, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Laboratorio de Fisiopatología Integrativa, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Leandro J Carreño
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe Melo-Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Translational Immunology Laboratory, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Claudia A Riedel
- Laboratorio de Endocrino-Inmunología, Centro de Investigación de Resiliencia a Pandemias, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| |
Collapse
|
15
|
Sorrentino G. Microenvironmental control of the ductular reaction: balancing repair and disease progression. Cell Death Dis 2025; 16:246. [PMID: 40180915 PMCID: PMC11968979 DOI: 10.1038/s41419-025-07590-4] [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: 10/16/2024] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
The ductular reaction (DR) is a dynamic adaptive cellular response within the liver, triggered by various hepatic insults and characterized by an expansion of dysmorphic biliary epithelial cells and liver progenitors. This complex response presents a dual role, playing a pivotal function in liver regeneration but, paradoxically, contributing to the progression of liver diseases, depending upon specific contextual factors and signaling pathways involved. This comprehensive review aims to offer a holistic perspective on the DR, focusing into its intricate cellular and molecular mechanisms, highlighting its pathological significance, and exploring its potential therapeutic implications. An up-to-date understanding of the DR in the context of different liver injuries is provided, analyzing its contributions to liver regeneration, inflammation, fibrosis, and ultimately carcinogenesis. Moreover, the review highlights the role of multiple microenvironmental factors, including the influence of extracellular matrix, tissue mechanics and the interplay with the intricate hepatic cell ecosystem in shaping the DR's regulation. Finally, in vitro and in vivo experimental models of the DR will be discussed, providing insights into how researchers can study and manipulate this critical cellular response. By comprehensively addressing the multifaceted nature of the DR, this review contributes to a more profound understanding of its pathophysiological role in liver diseases, thus offering potential therapeutic avenues for hepatic disorders and improving patient outcomes.
Collapse
Affiliation(s)
- Giovanni Sorrentino
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.
| |
Collapse
|
16
|
Jerab D, Blangero F, da Costa PCT, de Brito Alves JL, Kefi R, Jamoussi H, Morio B, Eljaafari A. Beneficial Effects of Omega-3 Fatty Acids on Obesity and Related Metabolic and Chronic Inflammatory Diseases. Nutrients 2025; 17:1253. [PMID: 40219010 PMCID: PMC11990730 DOI: 10.3390/nu17071253] [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: 02/27/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs) are known to help resolve inflammation through generation of anti-inflammatory eicosanoids and specialized pro-resolving mediators, including resolvins, protectins, and maresins. Through binding to the GPR120/FFAR4 receptor, their beneficial effects result from phospholipid membrane remodeling, impairment of inflammatory signaling molecules clustering, subsequent inhibition of NF-κB and inflammasome activation, and a reduction in oxidative stress. Obesity, a chronic inflammatory disease that contributes to metabolic disorders, is alleviated by n-3 PUFAs. In the adipose tissue (AT) of individuals with obesity, n-3 PUFAs counteract hypoxia, inhibit immune cell infiltration and AT inflammation, improve insulin sensitivity, and reduce fat mass. Beyond AT, n-3 PUFAs also alleviate other metabolic disorders such as metabolic-associated steatotic liver disease (MASLD), gut dysbiosis, and/or renal dysfunction. In cardiovascular disease (CVD), they are mainly recommended as a secondary prevention for patients with coronary heart disease risks. This review provides an in-depth analysis of the benefits of n-3 PUFAs in obesity and related metabolic diseases, examining both the mechanistic and clinical aspects. Additionally, it also explores the effects of n-3 PUFAs in obesity-related chronic inflammatory conditions, including inflammatory bowel disease, psoriasis, rheumatoid arthritis, osteoarthritis, and multiple sclerosis, by targeting specific pathophysiological mechanisms. Clinical applications and limitations of n-3 PUFAs are discussed based on findings from human clinical trials.
Collapse
Affiliation(s)
- Donia Jerab
- CarMeN Laboratory, Institut National de Recherche pour l’ Agriculture, l’ Alimentation et l’Environnement, UMR1397, Institut National de la Santé et de la Recherche Médicale, U 1060, Université Claude Bernard Lyon I, 69310 Pierre-Bénite, France (B.M.)
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis 1002, Tunisia;
| | - Ferdinand Blangero
- CarMeN Laboratory, Institut National de Recherche pour l’ Agriculture, l’ Alimentation et l’Environnement, UMR1397, Institut National de la Santé et de la Recherche Médicale, U 1060, Université Claude Bernard Lyon I, 69310 Pierre-Bénite, France (B.M.)
| | - Paulo César Trindade da Costa
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, Brazil (J.L.d.B.A.)
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, Brazil (J.L.d.B.A.)
| | - Rym Kefi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis 1002, Tunisia;
| | - Henda Jamoussi
- Research Unit “Obesity: Etiopathology and Treatment, UR18ES01”, Faculty of Medicine, Tunis El Manar University, Tunis 2092, Tunisia;
| | - Beatrice Morio
- CarMeN Laboratory, Institut National de Recherche pour l’ Agriculture, l’ Alimentation et l’Environnement, UMR1397, Institut National de la Santé et de la Recherche Médicale, U 1060, Université Claude Bernard Lyon I, 69310 Pierre-Bénite, France (B.M.)
| | - Assia Eljaafari
- CarMeN Laboratory, Institut National de Recherche pour l’ Agriculture, l’ Alimentation et l’Environnement, UMR1397, Institut National de la Santé et de la Recherche Médicale, U 1060, Université Claude Bernard Lyon I, 69310 Pierre-Bénite, France (B.M.)
- Department of Clinical Research, Hospices Civils de Lyon, 69002 Lyon, France
| |
Collapse
|
17
|
Tang Z, Jin L, Yang Y. The dual role of IL-17 in periodontitis regulating immunity and bone homeostasis. Front Immunol 2025; 16:1578635. [PMID: 40248692 PMCID: PMC12003107 DOI: 10.3389/fimmu.2025.1578635] [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: 02/18/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Periodontitis is a common dysbiotic bacteria-induced inflammatory disease characterized by alveolar bone resorption, leading to tooth loss. Interleukin-17 (IL-17) is a critical cytokine with dual roles in periodontium, which exerts the function of host defense, including neutrophil recruitment, phagocytosis, and mucosal immunity. However, excessive expression of IL-17 causes persistent chronic inflammation, local tissue breakdown, and bone loss. This review highlights the protective and pathological functions of IL-17 on immunity and bone homeostasis in inflammatory bone-related diseases. We also provide the latest findings with IL-17 knockout mice in periodontitis and highlight complex immune responses under various experimental models. This may provide a critical perception of inflammatory bone-related disease management using an immune-modulating strategy.
Collapse
Affiliation(s)
- Zhongyuan Tang
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Lijian Jin
- Division of Periodontology and Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yanqi Yang
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
18
|
Molitor TP, Hayashi G, Lin MY, Dunn CJ, Peterson NG, Poston RG, Kurnellas MP, Traver DA, Patel S, Akgungor Z, Leonardi V, Lewis C, Segales JS, Bennett DS, Truong AP, Dani M, Naphade S, Wong JK, McDermott AE, Kovalev SM, Ciaccio GL, Sadiq SA, Pei Z, Wood S, Rassoulpour A. Central TYK2 inhibition identifies TYK2 as a key neuroimmune modulator. Proc Natl Acad Sci U S A 2025; 122:e2422172122. [PMID: 40127268 PMCID: PMC12002270 DOI: 10.1073/pnas.2422172122] [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: 11/15/2024] [Accepted: 02/03/2025] [Indexed: 03/26/2025] Open
Abstract
GWAS have identified tyrosine kinase 2 (TYK2) variants in multiple inflammatory disorders, specifically a protective hypomorphic TYK2 allele (P1104A) in multiple sclerosis (MS). Impaired TYK2 signaling within the central nervous system (CNS) may impart the protective effects of TYK2 P1104A allele in MS. We deployed brain-penetrant TYK2 inhibitors (cTYK2i) alongside the peripherally restricted TYK2 inhibitor (pTYK2i; BMS-986165) to untangle the contributions of central TYK2 inhibition in diverse models of neuroinflammation. While pTYK2i had little impact, cTYK2i reduced clinical score, lymphoid cell infiltration, and cytokines/chemokines in experimental autoimmune encephalomyelitis (EAE). Microglial activation was attenuated in cTYK2i-treated EAE spinal cords and circulating neurofilament light (NfL) was reduced in plasma and cerebral spinal fluid (CSF). Additionally, cTYK2i was protective in an antibody-mediated mouse model of primary progressive MS (PPMS). Finally, we demonstrate TYK2 inhibition has a robust impact on a unique subset of activated astrocytes termed Interferon-Responsive-Reactive-Astrocytes (IRRA). The data presented herein identify a key role for CNS TYK2 signaling in regulating neuroinflammation and solidify TYK2 as a potential therapeutic target for MS.
Collapse
Affiliation(s)
- Tyler P. Molitor
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Genki Hayashi
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Mei-Yao Lin
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Carissa J. Dunn
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Robert G. Poston
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - David A. Traver
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Seona Patel
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Zeynep Akgungor
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Colizel Lewis
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Dylan S. Bennett
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Anh P. Truong
- Department of Chemistry, Neuron23, Inc., South San Francisco, CA94080
| | - Manjari Dani
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Swati Naphade
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Jamie K. Wong
- Tisch MS Research Center of New York, New York, NY10019
| | | | | | | | - Saud A. Sadiq
- Tisch MS Research Center of New York, New York, NY10019
| | - Zhonghua Pei
- Department of Chemistry, Neuron23, Inc., South San Francisco, CA94080
| | - Stephen Wood
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | |
Collapse
|
19
|
Vuppalanchi R, Liangpunsakul S. Editorial: Balancing the Yin and Yang of Alcohol-Associated Liver Disease-Integrating Pathophysiology, Liver-Directed Therapy and Addiction Management. Aliment Pharmacol Ther 2025; 61:1254-1255. [PMID: 40035413 DOI: 10.1111/apt.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025]
Affiliation(s)
- Raj Vuppalanchi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
20
|
Diaz LA, Morris S, Dave S, Kim SM, Sarik W, Richards L, Madamba E, Bettencourt R, Fulinara C, Pham T, Miller G, Carvalho-Gontijo Weber R, Momper JD, He F, Jain S, Jamieson C, Kisseleva T, Brenner D, Loomba R. Clinical Trial to Assess the Safety and Tolerability of Anti-IL 23 Monoclonal Antibody Guselkumab in Patients With Alcohol-Associated Liver Disease. Aliment Pharmacol Ther 2025; 61:1140-1151. [PMID: 39949265 DOI: 10.1111/apt.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND There are no FDA-approved therapies for alcohol-associated liver disease (ALD). Preclinical studies indicate that blocking IL-23/IL-17 signalling may reverse liver injury. Guselkumab, an IL-23-specific antibody approved for psoriasis, may be beneficial for ALD. AIMS We aimed to assess the safety and tolerability of guselkumab in patients with ALD. METHODS This phase-1 dose-escalation study included patients with ≥ 2 DSM-5 criteria for alcohol use disorder, significant steatosis (MRI-PDFF ≥ 8%) and MRE < 3.63 kPa (to exclude advanced disease). Guselkumab was given subcutaneously on Days 1 and 29 in 30, 70 or 100 mg dose cohorts. Primary endpoints were adverse events (AEs) and dose-limiting toxicity. RESULTS We enrolled 13 patients (three 30 mg, three 70 mg, and seven 100 mg). Eleven completed the study and two early discontinued in the 100 mg group. Of them, 77% were men, and the median age was 53 [IQR 49-61] years. The median MRI-PDFF and MRE were 18.4% [IQR 8.4%-34.0%] and 2.5 [2.2-2.6] kPa, respectively. The most frequent AEs were hyperuricemia (13%, mild only) and elevated lipase (11%, mild and moderate). There were no serious adverse events or significant variations in liver enzymes. There was a suppression of peripheral interleukin (IL)-17, IL-23, IL-1b and TNF-α in the 70 and 100 mg groups, and a significant decrease in alcohol consumption over time (AUDIT-C: 6 [3-7] vs. 5 [1-6], p = 0.023). CONCLUSIONS Guselkumab is safe in doses up to 100 mg and may reduce inflammation markers in ALD. These findings support further phase 2 studies to evaluate the efficacy of guselkumab in ALD, particularly in patients with severe phenotypes.
Collapse
Affiliation(s)
- Luis Antonio Diaz
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Sheldon Morris
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| | - Shravan Dave
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| | - Susy M Kim
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| | - Wathnita Sarik
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| | - Lisa Richards
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| | - Egbert Madamba
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| | - Ricki Bettencourt
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| | - Christian Fulinara
- Alpha Clinic, University of California San Diego, San Diego, California, USA
| | - Thuy Pham
- Alpha Clinic, University of California San Diego, San Diego, California, USA
| | - Grant Miller
- Department of Medicine, University of California San Diego School of Medicine, San Diego, California, USA
| | | | - Jeremiah D Momper
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California, USA
| | - Feng He
- Biostatistics Research Center, School of Public Health, University of California, San Diego, California, USA
| | - Sonia Jain
- Biostatistics Research Center, School of Public Health, University of California, San Diego, California, USA
| | - Catriona Jamieson
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| | - Tatiana Kisseleva
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
- Department of Surgery, University of California San Diego School of Medicine, San Diego, California, USA
| | - David Brenner
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
- Department of Medicine, University of California San Diego School of Medicine, San Diego, California, USA
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California, USA
| |
Collapse
|
21
|
Tian X, Peng F, Xiong X, Xu X, Zan Y, Wang X, Yu B, Liu Z, He X, Huang Z. Artemisinin analogues are effective in the treatment of psoriasis by targeting RORγt. Mol Immunol 2025; 180:11-22. [PMID: 39987640 DOI: 10.1016/j.molimm.2025.02.006] [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: 11/11/2024] [Revised: 02/02/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
Psoriasis is a chronic inflammatory skin autoimmune disease. Th17 cells, when pathologically activated, significantly contribute to the progression of psoriasis. The symptoms of this skin condition could be notably alleviated by targeting and suppressing the activity of these cells. Retinoic acid receptor-associated orphan nuclear hormone receptor γ-t (RORγt), a critical transcription factor in Th17 cells, emerges as a promising therapeutic target for autoimmune conditions which are mediated by the dysregulation of these cells. In this study, we designed and synthesised a series of artemisinin analogues based on the chemical structure of artemisinin, and screened 3 compounds, QHS-1, QHS-2, and QHS-3, with better inhibition efficiency of RORγt activity. We found that each of the three artemisinin analogues were demonstrated efficacy in curbing IMQ-induced skin inflammation and the abnormal proliferation of keratinocytes within the BALB/c mouse model of psoriasis. Our findings indicate that the three artemisinin analogues not only effectively mitigated skin inflammation and the abnormal proliferation of keratinocytes in the IMQ-induced psoriasis model of BALB/c mice but also curtailed the infiltration of immune cells and the production of pro-inflammatory cytokines in the dermis. Furthermore, these compounds modulated the cytokine expression profiles within Th17 cells. They exerted a suppressive effect on the activity of Th17 cells by targeting RORγt, thereby dampening the inflammatory response in the dorsal skin of the mice. This inhibition led to a reduction in the pathological proliferation of keratinocytes. In conclusion, our research underscores the promising therapeutic potential of artemisinin analogues in the treatment of psoriasis, offering a slate of candidate compounds which could pave the way for novel drug development in this field.
Collapse
Affiliation(s)
- Xuyan Tian
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Fanrong Peng
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Xiaoxiao Xiong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Xiaoting Xu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Yu Zan
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Xinran Wang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Bolan Yu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Zhonghua Liu
- Animal Experiment Center, South China Agricultural University, Guangzhou, China.
| | - Xixin He
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Zhaofeng Huang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| |
Collapse
|
22
|
Yoshida T, Takashima K, Mtali YS, Miyashita Y, Iwamoto A, Fukushima Y, Nakamura K, Oshiumi H. Regulation of IL-17A-mediated hypersensitivity by extracellular vesicles and lipid nanoparticles carrying miR-451a. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:651-665. [PMID: 40073105 DOI: 10.1093/jimmun/vkae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 12/04/2024] [Indexed: 03/14/2025]
Abstract
Extracellular vesicles (EVs), including exosomes, mediate intercellular communication by transporting functional molecules between donor cells and recipient cells, thereby regulating biological processes, such as immune responses. miR-451a, an immune regulatory microRNA, is highly abundant in circulating EVs; however, its precise physiological significance remains to be fully elucidated. Here, we demonstrate that miR-451a deficiency exacerbates delayed-type hypersensitivity (DTH) in mice. Notably, miR-451a knockout resulted in a significant increase in the number of interleukin (IL)-17A-expressing T helper 17 and γδ T cells infiltrating DTH-induced ear lesions. miR-451a deficiency also increased the number of γδ T cells in the secondary lymphoid tissues. Comprehensive analyses revealed that miR-451 deficiency promoted the expression of Rorc and γδ T cell-related genes following sensitization with allergens. Moreover, intravenous administration of wild-type EVs to miR-451a knockout mice increased cellular miR-451a levels in tissues and significantly attenuated the severity of DTH. Furthermore, synthetic lipid nanoparticles encapsulating miR-451a effectively mitigated DTH. Our findings indicate the importance of circulating miR-451a in the proliferation of γδ T cells and highlight the therapeutic potential of lipid nanoparticle-based microRNA delivery platforms for interventions in immune-related diseases.
Collapse
Affiliation(s)
- Takanobu Yoshida
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ken Takashima
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yohana S Mtali
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yusuke Miyashita
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Asuka Iwamoto
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshimi Fukushima
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
23
|
Hughes EP, Syage AR, Mehrabad EM, Lane TE, Spike BT, Tantin D. OCA-B promotes pathogenic maturation of stem-like CD4 + T cells and autoimmune demyelination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.29.569210. [PMID: 38076925 PMCID: PMC10705450 DOI: 10.1101/2023.11.29.569210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Stem-like T cells selectively contribute to autoimmunity, but the activities that promote their pathogenicity are incompletely understood. Here, we identify the transcription coregulator OCA-B as a driver of the pathogenic maturation of stem-like CD4 + T cell to promote autoimmune demyelination. Using two human multiple sclerosis (MS) datasets, we show that POU2AF1 , the gene encoding OCA-B, is elevated in CD4 + T cells from MS patients. We show that T cell-intrinsic OCA-B loss protects mice from experimental autoimmune encephalomyelitis (EAE) while preserving responses to viral CNS infection. In EAE models driven by antigen reencounter, OCA-B deletion nearly eliminates CNS infiltration, proinflammatory cytokine production and clinical disease. OCA-B-expressing CD4 + T cells of mice primed with autoantigen express an encephalitogenic gene program and preferentially confer disease. In a relapsing-remitting EAE model, OCA-B loss protects mice specifically at relapse. During remission, OCA-B promotes the expression of Tcf7 , Slamf6 , and Sell in proliferating CNS T cell populations. At relapse timepoints, OCA-B loss results in both the accumulation of an immunomodulatory CD4 + T cell population expressing Ccr9 and Bach2 , and loss of pro-inflammatory gene expression from Th17 cells. These results identify OCA-B as a driver of pathogenic CD4 + T cells.
Collapse
|
24
|
Zhang J, Shen M. The Role of IL-17 in Systemic Autoinflammatory Diseases: Mechanisms and Therapeutic Perspectives. Clin Rev Allergy Immunol 2025; 68:27. [PMID: 40074883 DOI: 10.1007/s12016-025-09042-5] [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] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Interleukin (IL)-17, a pro-inflammatory cytokine, plays a pivotal role in immune regulation by bridging innate and adaptive responses. Beyond its canonical involvement in T helper-17 cells-mediated immunity, IL-17 contributes significantly to the pathogenesis of systemic autoinflammatory diseases (SAIDs) including Familial Mediterranean Fever (FMF), nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-associated autoinflammatory diseases, and synovitis, acne, pustulosis, hyperostosis, and osteitis (SAPHO) syndrome. Dysregulated IL-17 signaling drives inflammasome activation, neutrophil recruitment, and chronic tissue inflammation. IL-17 inhibitors have demonstrated efficacy in refractory SAIDs, though challenges such as increased infection risks, paradoxical inflammatory reactions, and uncertainties regarding long-term safety persist. Currently, there is insufficient data to support the use of IL-17 inhibitors as first-line treatments, and their role in managing SAIDs is yet to be fully defined. This review highlights the mechanistic role of IL-17 in SAIDs and emerging therapeutic strategies, including IL-17-targeted monotherapies and combination approaches with IL-1 or tumor necrosis factor (TNF) inhibitors. Future research should focus on biomarker development, combination therapies, and long-term studies to optimize the safety and efficacy of IL-17-targeted therapies in SAIDs.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Min Shen
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
25
|
Ahmed S, Liu G, Sadiq A, Farooq U, Yang H, Yongbin L, Yiyu S, Xiaodong W, Jiang X. Integration of Immune Responses and Transcriptomic Signatures Reveals the Efficacy of Maternal Genetic Vaccination in a Pregnant Model and Its Neonates. Immunology 2025; 174:322-339. [PMID: 39762199 DOI: 10.1111/imm.13880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 02/07/2025] Open
Abstract
Maternal vaccination is essential for safeguarding both mother and foetus from infectious diseases. This study investigated the immunogenicity and efficacy of a maternal ORF-B2L genetic vaccine in a pregnant rat model, focusing on maternal-neonatal immune modulation, placental and neonatal spleen transcriptomics and the underlying mechanisms contributing to neonatal immune development. Female rats received intramuscular injections of either a gene vaccine (GV) containing 200 μg of recombinant ORF-B2L DNA and 50 μg of a subunit protein or an empty plasmid as a control. Results showed significantly higher levels of specific anti-B2L antibodies and Th1 and Th2 cytokine levels in both maternal and neonatal sera from the GV group compared to the control group (p < 0.05). Transcriptome analysis identified 1295 differentially expressed genes (DEGs) in the placenta and 998 DEGs in the neonatal spleen, with upregulated pathways associated with immune cell recruitment, cytokine signalling and hormone regulation in the GV group. Notably, upregulated DEGs such as TLR4, ESR1 and various cytokine/chemokine-related genes in the placenta suggest enhanced immune regulation and foetal protection. In the neonatal spleen, increased expression of IL-1β, IL-6, IL-10 and CD69 indicates enhanced T and B cell development and pathogen defence. The upregulation of IL-1β suggests a Th1 response, while elevated IL-10 indicates a potential Th2-biased immunity, reflecting a balanced Th1/Th2 response that is crucial for effective adaptive immunity. Overall, maternal ORF-B2L genetic vaccination induces a robust immune response, enhancing maternal-foetal protection and shaping neonatal immune responses, offering valuable insights for optimizing maternal vaccination strategies.
Collapse
Affiliation(s)
- Sohail Ahmed
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Guiqiong Liu
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Amber Sadiq
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Umar Farooq
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Huiguo Yang
- Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Liu Yongbin
- College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Sha Yiyu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wang Xiaodong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Xunping Jiang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
26
|
Camard L, Stephen T, Yahia-Cherbal H, Guillemot V, Mella S, Baillet V, Lopez-Maestre H, Capocefalo D, Cantini L, Leloup C, Marsande J, Garro K, Sienes Bailo J, Dangien A, Pietrosemoli N, Hasan M, Wang H, Eckle SB, Fourie AM, Greving C, Joyce-Shaikh B, Parker R, Cua DJ, Bianchi E, Rogge L. IL-23 tunes inflammatory functions of human mucosal-associated invariant T cells. iScience 2025; 28:111898. [PMID: 40008359 PMCID: PMC11850163 DOI: 10.1016/j.isci.2025.111898] [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: 06/05/2024] [Revised: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
IL-23 signaling plays a key role in the pathogenesis of chronic inflammatory and infectious diseases, yet the cellular targets and signaling pathways affected by this cytokine remain poorly understood. We show that IL-23 receptors are expressed on the large majority of human mucosal-associated invariant T (MAIT), but not of conventional T cells. Protein and transcriptional profiling at the population and single cell level demonstrates that stimulation with IL-23 or the structurally related cytokine IL-12 drives distinct functional profiles, revealing a high level of plasticity of MAIT cells. IL-23, in particular, affects key molecules and pathways related to autoimmunity and cytotoxic functions. Integrated analysis of transcriptomes and chromatin accessibility, supported by CRISPR-Cas9 mediated deletion, shows that AP-1 transcription factors constitute a key regulatory node of the IL-23 pathway in MAIT cells. In conclusion, our findings indicate that MAIT cells are key mediators of IL-23 functions in immunity to infections and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Laetitia Camard
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Tharshana Stephen
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
- scBiomarkers, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Hanane Yahia-Cherbal
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Vincent Guillemot
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Sébastien Mella
- scBiomarkers, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Victoire Baillet
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Hélène Lopez-Maestre
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Daniele Capocefalo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics Group, 75015 Paris, France
| | - Laura Cantini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics Group, 75015 Paris, France
| | - Claire Leloup
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Julie Marsande
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Katherine Garro
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Juan Sienes Bailo
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Ambre Dangien
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
- Department of Dermatology, Hôpital Cochin, AP-HP, AP-HP Centre-Université de Paris, 75014 Paris, France
| | - Natalia Pietrosemoli
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Milena Hasan
- scBiomarkers, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Huimeng Wang
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sidonia B.G. Eckle
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Anne M. Fourie
- Janssen Research & Development, LLC, San Diego, CA 92121, USA
| | - Carrie Greving
- Janssen Research & Development, LLC, San Diego, CA 92121, USA
| | | | - Raphaelle Parker
- Janssen Research & Development, Janssen-Cilag, 92130 Issy les Moulineaux, France
| | - Daniel J. Cua
- Janssen Research & Development, LLC, Spring House, PA 19002, USA
| | - Elisabetta Bianchi
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Lars Rogge
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| |
Collapse
|
27
|
Kuklina EM. Mechanisms of Glucocorticoid Resistance in Nonclassical T Helper Populations Th17.1/Ex-Th17. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:188-199. [PMID: 40254398 DOI: 10.1134/s0006297924604222] [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: 11/25/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 04/22/2025]
Abstract
The nonclassical population of Th1-polarized Th17 lymphocytes (Th17.1/ex-Th17) is currently in the focus of researchers' attention. These cells possess a unique proinflammatory potential and ability to penetrate blood-tissue barriers and play a key role in the pathogenesis of many inflammatory diseases, primarily autoimmune ones. Th1-polarized Th17 lymphocytes prevail in the autoimmune lesion foci and are considered to be a promising therapeutic target in these pathologies. At the same time, recent studies have shown another distinctive feature of Th1-polarized Th17 - their selective resistance to glucocorticoids. Since glucocorticoids are the first-line drugs for the treatment of the autoimmune disease exacerbation, understanding the causes of this phenomenon is crucial for predicting patients' response to therapy and improving the treatment effectiveness. This review analyzes the mechanisms of drug resistance of Th1-polarized Th17 cells, compares these mechanisms with those typical of nonpathogenic classical Th17 cells, and discusses the role of glucocorticoid resistance in the body's response to glucocorticoid therapy.
Collapse
Affiliation(s)
- Elena M Kuklina
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia.
| |
Collapse
|
28
|
Zhang P, Pei B, Yi C, Akanyibah FA, Mao F. The role of suppressor of cytokine signaling 3 in inflammatory bowel disease and its associated colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167578. [PMID: 39571630 DOI: 10.1016/j.bbadis.2024.167578] [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: 10/01/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024]
Abstract
Inflammatory bowel disease (IBD) and colorectal cancer (CRC), as two of the major human intestinal diseases, provide challenges for the medical field. Suppressor of cytokine signaling 3 (SOCS3), a protein molecule that negatively regulates cytokine signaling through multiple pathways, is involved in the regulation of various inflammatory diseases and tumors. In IBD, SOCS3 acts on a variety of cells to repair mucosal damage and balance the immune response, including epithelial cells, macrophages, dendritic cells, neutrophils, and T cells. In CRC, SOCS3 is inextricably linked to tumor cell proliferation, invasion, metastasis, and drug resistance. Therefore, it is crucial to systematically investigate the pathogenic involvement of SOCS3 in IBD and CRC. This article reviews the mechanisms and pathways by which SOCS3 is involved in the inhibition of IBD and the mitigation of CRC, and details the therapeutic options for targeting SOCS3.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, Jiangsu, PR China; Institute of Hematology, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian 223800, Jiangsu, PR China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang 212028, PR China
| | - Francis Atim Akanyibah
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, Jiangsu, PR China
| | - Fei Mao
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, Jiangsu, PR China; Institute of Hematology, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China.
| |
Collapse
|
29
|
Vieujean S, Jairath V, Peyrin-Biroulet L, Dubinsky M, Iacucci M, Magro F, Danese S. Understanding the therapeutic toolkit for inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-024-01035-7. [PMID: 39891014 DOI: 10.1038/s41575-024-01035-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 02/03/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing ulcerative colitis and Crohn's disease, is a group of chronic, immune-mediated disorders of the gastrointestinal tract that present substantial clinical challenges owing to their complex pathophysiology and tendency to relapse. A treat-to-target approach is recommended, involving iterative treatment adjustments to achieve clinical response, reduce inflammatory markers and achieve long-term goals such as mucosal healing. Lifelong medication is often necessary to manage the disease, maintain remission and prevent complications. The therapeutic landscape for IBD has evolved substantially; however, a ceiling on therapeutic efficacy remains and surgery is sometimes required (owing to uncontrolled disease activity or complications). Effective IBD management involves comprehensive care, including medication adherence and a collaborative clinician-patient relationship. This Review discusses current therapeutic options for IBD, detailing mechanisms of action, efficacy, safety profiles and guidelines for use of each drug class. We also explore emerging therapies and the role of surgery. Additionally, the importance of a multidisciplinary team and personalized care in managing IBD is emphasized, advocating for patient empowerment and involvement in treatment decisions. By synthesizing current knowledge and emerging trends, this Review aims to equip healthcare professionals with a thorough understanding of therapeutic options for IBD, enhancing informed, evidence-based decisions in clinical practice.
Collapse
Affiliation(s)
- Sophie Vieujean
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, Liège, Belgium
- Department of Gastroenterology, INFINY Institute, CHRU Nancy, Vandœuvre-lès-Nancy, France
| | - Vipul Jairath
- Division of Gastroenterology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, INFINY Institute, CHRU Nancy, Vandœuvre-lès-Nancy, France
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Marla Dubinsky
- Department of Paediatrics, Susan and Leonard Feinstein IBD Center, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Marietta Iacucci
- APC Microbiome Ireland, College of Medicine and Health, University College of Cork, Cork, Ireland
| | - Fernando Magro
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, Milano, Italy.
| |
Collapse
|
30
|
Smith KJ, Lim Z, Vermeren S, Miron VE, Dimeloe S, Davidson DJ, Williams A, Gwyer Findlay E. Proteomic analysis reveals dysregulation of peripheral blood neutrophils in patients with Multiple Sclerosis. Clin Exp Immunol 2025; 219:uxae115. [PMID: 39817476 PMCID: PMC12124191 DOI: 10.1093/cei/uxae115] [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: 06/10/2024] [Revised: 10/13/2024] [Accepted: 12/19/2024] [Indexed: 01/18/2025] Open
Abstract
Multiple Sclerosis (MS) is a complex auto-inflammatory disease affecting the brain and spinal cord, which results in axonal de-myelination and symptoms including fatigue, pain, and difficulties with vision and mobility. The involvement of the immune system in the pathology of MS is well established, particularly the adaptive T cell response, and there has been a particular focus on the IL-17-producing subset of Th17 cells and their role in driving disease. However, the importance of innate immune cells has not been so well characterized. Here we focussed on neutrophils, which are innate immune cells and rapid responders to inflammation, and which have recently been linked to other chronic autoimmune conditions. Multiple strands of evidence in patients with MS and in mice with the experimental autoimmune encephalomyelitis MS model suggest neutrophils may play a role in driving MS inflammation. Here, we performed proteomic analysis on neutrophils from patients with MS and healthy donors, revealing striking differences. In particular, granule proteins were significantly more abundant in the MS neutrophils compared to the healthy controls, with a particular overabundance of proteins in primary and secondary granules. In addition, members of the MAVS signalling pathway were differently regulated compared to healthy donor cells. Finally, we find that MS neutrophils do not suppress T cell activation equivalently to healthy neutrophils, and in particular are unable to suppress expression of CD161 on the T cells, indicative of a suppression of Th17 differentiation. We propose that neutrophil dysregulation in MS may contribute to dysfunctional T cell responses.
Collapse
Affiliation(s)
- Katie J Smith
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Zachary Lim
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Sonja Vermeren
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Keenan Research Centre for Biomedical Sciences at St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sarah Dimeloe
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Donald J Davidson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Anna Williams
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, University of Edinburgh, UK
| | - Emily Gwyer Findlay
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- School of Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
31
|
Henry-Ojo HO, Liu F, Narayanan SP. Targeting SMOX Preserves Optic Nerve Myelin, Axonal Integrity, and Visual Function in Multiple Sclerosis. Biomolecules 2025; 15:158. [PMID: 40001462 PMCID: PMC11853291 DOI: 10.3390/biom15020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
Multiple sclerosis (MS) is a highly disabling chronic neurological condition affecting young adults. Inflammation, demyelination, and axonal damage are key pathological features of MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Our previous work demonstrated that inhibiting spermine oxidase (SMOX) with MDL72527, a selective irreversible pharmacological inhibitor, significantly reduced clinical symptoms, retinal ganglion cell (RGC) loss, and optic nerve inflammation in EAE mice. The present study explored the broader therapeutic potential of SMOX inhibition, focusing on myelin preservation, axonal integrity, and visual function in the EAE model. Electron microscopy of optic nerve cross-sections showed significant preservation of myelin thickness and axonal integrity due to SMOX inhibition. The quantitative assessment showed that g-ratio and axon count metrics were significantly improved in MDL72527-treated EAE mice compared to their vehicle-treated counterparts. Immunofluorescence studies confirmed these findings, showing increased preservation of myelin and axonal proteins in MDL72527-treated EAE mice compared to the vehicle-treated group. Functional assessment studies (Electroretinography) demonstrated significant improvement in RGC function and axonal conduction in EAE mice treated with MDL72527. Furthermore, SMOX inhibition downregulated the expression of galectin3 (Gal3), a mediator of neuroinflammation, indicating Gal3's role in SMOX-mediated neuroprotection. This study provides compelling evidence for the potential of SMOX inhibition as a therapeutic strategy in multiple sclerosis and other demyelinating disorders.
Collapse
Affiliation(s)
- Harry O. Henry-Ojo
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30907, USA; (H.O.H.-O.); (F.L.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30907, USA
| | - Fang Liu
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30907, USA; (H.O.H.-O.); (F.L.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30907, USA
| | - S. Priya Narayanan
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30907, USA; (H.O.H.-O.); (F.L.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30907, USA
| |
Collapse
|
32
|
Shi W, Zhao Z, Zhai Y, Ye X, Xu F. Adverse events associated with IL-23 and IL-12/23 inhibitors in the clinical management of psoriasis: a comprehensive pharmacovigilance analysis. BMC Pharmacol Toxicol 2025; 26:11. [PMID: 39833957 PMCID: PMC11748260 DOI: 10.1186/s40360-025-00837-y] [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: 10/30/2023] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Interleukin-23 (IL-23) inhibitors and the IL-12/23 inhibitor ustekinumab constitute a pivotal class of therapeutic agents employed in the clinical management of Psoriasis, a chronic immune-mediated skin disorder. Notwithstanding their therapeutic efficacy, concerns have arisen due to the emergence of multiple adverse events (AEs) associated with their usage. This study aims to provide a comprehensive examination of the distribution and characteristics of these AEs concerning IL-23 and IL-12/23 inhibitors, with a specific focus on guselkumab, tildrakizumab, risankizumab, and ustekinumab. METHODS In this research endeavor, we conducted an extensive analysis of data extracted from the FDA Adverse Event Reporting System (FAERS), spanning the timeframe from January 1, 2014, to September 30, 2022. To identify potential signals of AEs, we rigorously applied disproportionality analysis, utilizing both reporting odds ratio (ROR) and information component (IC) metrics. A signal was considered present when the lower limit of the 95% confidence interval (CI) for ROR (ROR025) exceeded one or when IC (IC025) surpassed zero, with a minimum requirement of three or more reported cases. RESULTS Our investigation encompassed a substantial dataset, comprising a total of 41,408,408 reports detailing drug-AE associations and involving 13,271,168 individuals. Among these, 704, 13,164, and 11,399 patients were identified as users of the IL-23 inhibitors tildrakizumab, guselkumab, and risankizumab, respectively, while 62,853 patients were identified as users of the IL-12/23 inhibitor ustekinumab. The analysis revealed the presence of 8, 20, 107, and 115 signals for these respective drugs. Significantly, the System Organ Class (SOC) exhibiting the highest incidence was "infections and infestations," with documented occurrences in tildrakizumab (6/8), guselkumab (5/20), ustekinumab (50/107), and risankizumab (25/115). CONCLUSION Our pharmacovigilance analysis has brought to light a substantial frequency of AEs linked to IL-23 and IL-12/23 inhibitors. These findings underscore the pivotal role of IL-23 and IL-12/23 inhibitors in modulating immune function and raise concerns regarding their potential to heighten susceptibility to infections and malignancies. However, limitations inherent to the FAERS database, including underreporting, lack of denominator data, potential duplicate records, and inability to confirm causality, should be acknowledged of particular significance is risankizumab, which, despite having fewer reported cases and a later market introduction compared to ustekinumab, exhibited a higher incidence of AEs. These results emphasize the necessity for ongoing vigilance, further investigation, and a reevaluation of the safety profile of IL-23 and IL-12/23 inhibitors in the clinical management of Psoriasis.
Collapse
Affiliation(s)
- Wentao Shi
- Clinical Research Unit, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyi Zhao
- Rehabilitation Center, The First Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Yinghong Zhai
- Clinical Research Unit, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofei Ye
- Department of Health Statistics, Naval Medical University, Shanghai, China.
| | - Feng Xu
- Clinical Research Unit, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
33
|
Collins M, Pehrson R, Grindebacke H, Leffler A, Ramnegård M, Rannikmäe H, Krutrök N, Yrlid L, Pollard C, Dainty I, Narjes F, von Berg S, Llinas A, Malmberg A, McPheat J, Hansson E, Bäck E, Bernström J, Hansson TG, Keeling D, Jirholt J. RORγt inverse agonists demonstrating a margin between inhibition of IL-17A and thymocyte apoptosis. PLoS One 2025; 20:e0317090. [PMID: 39820614 PMCID: PMC11737796 DOI: 10.1371/journal.pone.0317090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 12/21/2024] [Indexed: 01/19/2025] Open
Abstract
Multiple genetic associations suggest a causative relationship between Th17-related genes coding for proteins, such as IL-17A, IL-23 and STAT3, and psoriasis. Further support for this link comes from the findings that neutralizing antibodies directed against IL-17A, IL-17RA and IL-23 are efficacious in diseases like psoriasis, psoriatic arthritis and ankylosing spondylitis. RORγt is a centrally positioned transcription factor driving Th17 polarization and cytokine secretion and modulation of RORγt may thus provide additional benefit to patients. However, RORγt also plays a role in the normal development of T cells in the thymus and genetic disruption of RORγt in the mouse leads to the development of lymphoma originating in the thymus. Whilst it is not established that down-regulation of RORγt activity would lead to the same consequence in humans, further understanding of the thymus effects is desirable to support progress of this target as a potential treatment of Th17-driven disease. Herein we present the characterisation of recently disclosed RORγt inverse agonists demonstrating target engagement and efficacy in vitro and in vivo against Th17 endpoints but requiring higher concentrations in vitro to affect thymocyte apoptosis.
Collapse
Affiliation(s)
- Mia Collins
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rikard Pehrson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Hanna Grindebacke
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Agnes Leffler
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marie Ramnegård
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Helena Rannikmäe
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Nina Krutrök
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Linda Yrlid
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Charlotte Pollard
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ian Dainty
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Frank Narjes
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stefan von Berg
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Antonio Llinas
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Malmberg
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jane McPheat
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eva Hansson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Bäck
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jenny Bernström
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Thomas G. Hansson
- Projects, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - David Keeling
- Projects, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Johan Jirholt
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
34
|
Li J, Mao N, Wang Y, Deng S, Chen K. Novel insights into the ROCK-JAK-STAT signaling pathway in upper respiratory tract infections and neurodegenerative diseases. Mol Ther 2025; 33:32-50. [PMID: 39511889 PMCID: PMC11764622 DOI: 10.1016/j.ymthe.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024] Open
Abstract
Acute upper respiratory tract infections are a major public health issue, with uncontrolled inflammation triggered by upper respiratory viruses being a significant cause of patient deterioration or death. This study focuses on the Janus kinase-signal transducer and activator of transcription Rho-associated coiled-coil containing protein kinase (JAK-STAT-ROCK) signaling pathway, providing an in-depth analysis of the interplay between uncontrolled inflammation after upper respiratory tract infections and the development of neurodegenerative diseases. It offers a conceptual framework for understanding the lung-brain-related immune responses and potential interactions. The relationship between the ROCK-JAK-STAT signaling pathway and inflammatory immunity is a complex and multi-layered research area and exploring potential common targets could open new avenues for the prevention and treatment of related inflammation.
Collapse
Affiliation(s)
- Jiaxuan Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Naihui Mao
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| | - Shuli Deng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China.
| |
Collapse
|
35
|
Ren YK, Ren L, Sun W, Liu HY, Guo SP. Paradoxical Behçet's Disease after Ixekizumab: A Case Report and Literature Review. Indian J Dermatol 2025; 70:42-46. [PMID: 39896301 PMCID: PMC11784972 DOI: 10.4103/ijd.ijd_719_24] [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: 08/01/2024] [Accepted: 09/01/2024] [Indexed: 02/04/2025] Open
Abstract
Behçet's disease is a chronic inflammatory condition that affects multiple organs and systems. It is characterized by recurrent oral and genital ulcers. A previous study reported that the IL-17A inhibitor secukinumab can improve the skin and mucosal manifestations in patients with refractory Behçet's disease. Additionally, secukinumab has been shown to effectively improve neurological symptoms when administered for Behçet's disease. However, we report a case where Behçet's disease occurred after the treatment of psoriasis with the IL-17A inhibitor ixekizumab. To summarize its clinical characteristics and treatment experience, we consulted relevant domestic and international literature and conducted a literature review. We concluded that anti-IL-17A treatment may lead to the development of Behçet's disease. The reported cases are more likely to occur in middle-aged men with varying onset times. The main manifestations include oral and vulvar mucosal ulcers. Furthermore, the gut microbiota may be involved in paradoxical Behçet's disease.
Collapse
Affiliation(s)
- Yue-Kang Ren
- From the Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ling Ren
- Department of Dermatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wen Sun
- From the Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hong-Ye Liu
- Department of Dermatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shu-Ping Guo
- Department of Dermatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
36
|
Tashiro H, Kuwahara Y, Kurihara Y, Takahashi K. Molecular mechanisms and clinical impact of biologic therapies in severe asthma. Respir Investig 2025; 63:50-60. [PMID: 39642687 DOI: 10.1016/j.resinv.2024.11.015] [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: 09/30/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Severe asthma is a critical condition for patients with asthma, characterized by frequent exacerbations, decreased pulmonary function, and unstable symptoms related to asthma. Consequently, the administration of systemic corticosteroids, which cause secondary damage because of their adverse effects, is considered. Recently, several types of molecular-targeted biological therapies have become available for patients with severe asthma, and they have a capacity to improve the pathophysiology of severe asthma. However, several clinical reports indicate that the effects differ depending on the biological targets of asthma in individual patients. In this review, the molecular mechanisms and clinical impact of biologic therapies in severe asthma are described. In addition, molecules targeted by possible future biologics are also addressed. Better understanding of the mechanistic basis for the role of biologics in severe asthma could lead to new therapeutic options for these patients.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kuwahara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan.
| |
Collapse
|
37
|
Park DJ, Joo YB, Nam E, Lee J, Bang SY, Lee HS, Bae SC. Exploring potential multiple molecular biomarkers that predict treatment response in patients with lupus nephritis. Sci Rep 2024; 14:31422. [PMID: 39733104 PMCID: PMC11682382 DOI: 10.1038/s41598-024-83057-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Limited knowledge exists regarding biomarkers that predict treatment response in Lupus nephritis (LN). We aimed to identify potential molecular biomarkers to predict treatment response in patients with LN. We enrolled 66 patients with active LN who underwent renal biopsy upon enrollment. Serum and urine samples were collected longitudinally, and we measured 12 biomarkers in each sample using a multiplex immunofluorescence assay. These biomarkers included monocyte chemoattractant protein-1 (MCP-1), interferon gamma-induced protein 10 (IP-10), interferon-γ (IFN-γ), interleukin 6 (IL-6), interleukin 16 (IL-16), interleukin 17 (IL-17), interleukin 23 (IL-23), tumor necrosis factor receptor II (TNF-RII), vascular cell adhesion molecule 1 (VCAM-1), retinol-binding protein 4 (RBP 4), vitamin D binding protein (VDBP), and neutrophil gelatinase-associated lipocalin (NGAL). Patients were categorized into two groups based on their 1-year treatment response to Mycophenolate mofetil (MMF)-based therapy: 50 responders and 16 non-responders. Only urine IL-17 (uIL-17) showed baseline level differences between the two groups, with higher in responders. In ROC curve analyses assessing the predictive performance of biomarkers, baseline uIL-17 and changes in uIL-6 and uIL-23 levels at 3 months could predict the 1-year treatment response, showing AUC values of 0.70 (95% CI 0.54-0.87), 0.70 (0.54-0.86), and 0.71 (0.57-0.85), respectively. Combining uIL-6 and uIL-23 into a model improved predictability, achieving an AUC of 0.75 (0.61-0.90). Baseline uIL-17 levels and early changes in uIL-6 and uIL-23 could serve as potential biomarkers to predict 1-year treatment response in lupus nephritis patients receiving MMF-based therapy.
Collapse
Affiliation(s)
- Dae Jin Park
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea
| | - Young Bin Joo
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea
| | - Eunwoo Nam
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea
| | - Jiyoung Lee
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea
| | - So-Young Bang
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea
| | - Hye-Soon Lee
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea.
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea.
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea.
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea.
| |
Collapse
|
38
|
Zhu W, Revu S, Chen C, Dahl M, Ramkumar A, Kelly C, McGeachy MJ, Xia Z. Aging-dependent Change in Th17 and Cytokine Response in Multiple Sclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.17.24304425. [PMID: 39763560 PMCID: PMC11702752 DOI: 10.1101/2024.03.17.24304425] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic autoimmune disease damaging the central nervous system. Diminished inflammatory disease activity (DA) as people with MS (pwMS) age motivated randomized clinical trials assessing disease-modifying therapy (DMT) discontinuation in older pwMS given the concern for risks outweighing benefits. This study aims to examine whether peripheral production of Myelin Basic Protein (MBP)-driven cytokine responses mediate the aging-associated decline in MS inflammatory DA. METHODS We included the clinical data of 669 adult pwMS between 2017 and 2022 who enrolled in a clinic-based prospective cohort. From a subset of 80 participants, we isolated fresh peripheral blood mononuclear cells (PBMCs) and cultured with 50μg/ml of MBP (or heat-killed Candida) for 24 hours. We assayed cell culture supernatants for interleukin 17 (IL-17) and interferon gamma (IFN-γ) using Enzyme-Linked Immunosorbent Assay and a subset of the supernatant samples using a commercial human cytokine/chemokine array. We examined the associations between age and annualized relapse rate (ARR) as well as between age and MBP-stimulated cytokine production (by cultured PBMC) using covariate-adjusted linear regressions. We performed mediation analyses to determine the extent to which MBP-driven cytokine response drives the association between age and ARR. RESULTS Among 669 pwMS (mean age 51.7±12.7 years, 80.7% women, 89.4% non-Hispanic White), ARR declined with age (β=-0.003, p<0.001). Among the subgroup of 80 pwMS whose cultured PBMCs underwent ex vivo MBP stimulation, IL-17 production declined with age in women (β=-0.27, p=0.04) but not men (β=-0.1, p=0.73). MBP-driven IL-17 response partially mediated the association between older age and lower ARR (24.7% in women, 15.3% in men). In exploratory analyses, older pwMS (≥50 years) had marginally lower (IL-4, MCP-2, MCP-3, PDGF-AA, PDGF-AB/BB) and higher (Fractalkine, MDC) concentrations of several cytokines than younger pwMS (<50 years), while certain cytokines (MCP-2, MDC) mediated whereas others negated the effect of age on ARR. CONCLUSION Diminished peripheral IL-17 response as a potential biological mechanism underlying the aging-dependent decline in MS inflammatory DA warrants further investigation.
Collapse
|
39
|
Kot A, Koszewska D, Ochman B, Świętochowska E. Clinical Potential of Misshapen/NIKs-Related Kinase (MINK) 1-A Many-Sided Element of Cell Physiology and Pathology. Curr Issues Mol Biol 2024; 46:13811-13845. [PMID: 39727954 PMCID: PMC11727420 DOI: 10.3390/cimb46120826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Misshapen/NIKs-related kinase (MINK) 1 belongs to the mammalian germinal center kinase (GCK) family. It contains the N-terminal, conserved kinase domain, a coiled-coil region, a proline-rich region, and a GCK, C-terminal domain with the Citron-NIK-Homology (CNH) domain. The kinase is an essential component of cellular signaling pathways, which include Wnt signaling, JNK signaling, pathways engaging Ras proteins, the Hippo pathway, and STRIPAK complexes. It thus contributes to regulating the cell cycle, apoptosis, cytoskeleton organization, cell migration, embryogenesis, or tissue homeostasis. MINK1 plays an important role in immunological responses, inhibiting Th17 and Th1 cell differentiation and regulating NLRP3 inflammasome function. It may be considered a link between ROS and the immunological system, and a potential antiviral target for human enteroviruses. The kinase has been implicated in the pathogenesis of sepsis, rheumatoid arthritis, asthma, SLE, and more. It is also involved in tumorigenesis and drug resistance in cancer. Silencing MINK1 reduces cancer cell migration, suggesting potential for new therapeutic approaches. Targeting MINK1 could be a promising treatment strategy for patients insensitive to current chemotherapies, and could improve their prognosis. Moreover, MINK1 plays an important role in the nervous system and the cardiovascular system development and function. The modulation of MINK1 activity could influence the course of neurodegenerative diseases, including Alzheimer's disease. Further exploration of the activity of the kinase could also help in gaining more insight into factors involved in thrombosis or congenital heart disease. This review aims to summarize the current knowledge on MINK1, highlight its therapeutic and prognostic potential, and encourage more studies in this area.
Collapse
Affiliation(s)
| | | | | | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland; (A.K.); (D.K.); (B.O.)
| |
Collapse
|
40
|
Warren RB, Pavlovsky L, Costanzo A, Bukhalo M, Korman NJ, Huang YH, Kokolakis G, Pinter A, Ibrahim N, Zheng Y, Drogaris L, Stakias V, Soliman AM, Rubant S, Thaçi D. Efficacy and Safety of Risankizumab in Patients with Psoriasis Showing Suboptimal Response to Secukinumab or Ixekizumab: Results from a Phase 3b, Open-Label, Single-Arm (aIMM) Study. Dermatol Ther (Heidelb) 2024; 14:3273-3290. [PMID: 39516454 PMCID: PMC11604970 DOI: 10.1007/s13555-024-01292-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Risankizumab has demonstrated superior efficacy compared to other psoriasis treatments, including secukinumab, adalimumab, and ustekinumab; switching to risankizumab from other psoriasis treatments has shown superior clinical and quality of life (QoL) outcomes. We evaluated the efficacy and safety of directly switching patients with moderate-to-severe plaque psoriasis and a suboptimal response to interleukin (IL)-17 inhibitors (secukinumab or ixekizumab) to risankizumab. METHODS This 52-week, phase 3b study enrolled patients (≥ 18 years) with moderate-to-severe plaque psoriasis who had previously been treated with the recommended dose of secukinumab or ixekizumab for ≥ 6 months but did not achieve an optimal response (static Physician's Global Assessment [sPGA] 2/3; body surface are [BSA] 3- < 10%). Patients received subcutaneous risankizumab (150 mg) without washout. The primary endpoint was the proportion of patients achieving sPGA of 0/1 at week 16. Secondary endpoints included sPGA 0/1 at week 52, sPGA 0, Dermatology Life Quality Index (DLQI) 0/1, and Psoriasis Symptoms Scale (PSS) 0 at weeks 16 and 52. Safety was monitored throughout the study. RESULTS The study included 244 patients. sPGA 0/1 was achieved by 57.4% and 62.3% at week 16 and 52. At week 16, sPGA 0, DLQI 0/1, and PSS 0 were achieved by 20.5%, 40.2%, and 20.9%, respectively. At week 52, these proportions increased to 27.1% for sPGA 0, 47.2% for DLQI 0/1, and 27.5% for PSS 0. Most frequently reported adverse events (reported in ≥ 5% of patients) in risankizumab-treated patients were COVID-19 infection (8.6%) and nasopharyngitis (5.7%). No new safety signals were observed. CONCLUSIONS Directly switching to risankizumab improved outcomes and QoL in patients with moderate-to-severe psoriasis who had suboptimal responses to anti-IL-17 inhibitors (secukinumab or ixekizumab). The safety results are consistent with previously reported safety of risankizumab. This study supports the efficacy of risankizumab in patients previously treated with biologics, including IL-17 inhibitors, and suggests a direct switch to risankizumab for improved clinical outcomes and QoL. CLINICAL TRIALS ClinicalTrials.gov identifier: NCT04102007.
Collapse
Affiliation(s)
- Richard B Warren
- The Dermatology Centre, Northern Care Alliance NHS Foundation Trust, Salford Royal NHS Foundation Trust, Irving Building, Salford, Manchester, M6 8HD, UK.
- NIHR Manchester Biomedical Research Center, Manchester University NHS Foundation Trust, Manchester Academic Health Science Center, Manchester, UK.
| | - Lev Pavlovsky
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Antonio Costanzo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Neil J Korman
- Department of Dermatology, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH, USA
| | - Yu-Huei Huang
- Department of Dermatology, Linkou Branch, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Georgios Kokolakis
- Psoriasis Research and Treatment Center, Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Pinter
- Department of Dermatology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | | | | | | | | | | | | | - Diamant Thaçi
- Institute and Comprehensive Center Inflammation Medicine, University of Lübeck, Lübeck, Germany
| |
Collapse
|
41
|
Halder N, Yadav S, Lal G. Neuroimmune communication of the cholinergic system in gut inflammation and autoimmunity. Autoimmun Rev 2024; 23:103678. [PMID: 39500481 DOI: 10.1016/j.autrev.2024.103678] [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: 10/01/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Neuroimmune communication in the body forms a bridge between two central regulatory systems of the body, i.e., nervous and immune systems. The cholinergic system is a crucial modulatory neurotransmitter in the central and peripheral nervous system. It includes the neurotransmitter acetylcholine (ACh), the enzyme required for the synthesis of ACh (choline acetyltransferase, ChAT), the enzyme required for its degradation (acetylcholinesterase, AChE), and cholinergic receptors (Nicotinic acetylcholine receptors and muscarinic acetylcholine receptors). The cholinergic system in neurons is well known for its role in cognitive function, sensory perception, motor control, learning, and memory processes. It has been shown that the non-neuronal cholinergic system (NNCS) is present in various tissues and immune cells and forms a neuroimmune communications system. In the present review, we discussed the NNCS on immune cells, its role in homeostasis and inflammatory reactions in the gut, and how it can be exploited in treating inflammatory responses.
Collapse
Affiliation(s)
- Namrita Halder
- Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), SPPU campus, Ganeshkhind, Pune, MH-411007, India
| | - Sourabh Yadav
- Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), SPPU campus, Ganeshkhind, Pune, MH-411007, India
| | - Girdhari Lal
- Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), SPPU campus, Ganeshkhind, Pune, MH-411007, India.
| |
Collapse
|
42
|
Okamoto M, Kuratani A, Okuzaki D, Kamiyama N, Kobayashi T, Sasai M, Yamamoto M. IFN-γ-induced Th1-Treg polarization in inflamed brains limits exacerbation of experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 2024; 121:e2401692121. [PMID: 39560646 PMCID: PMC11621829 DOI: 10.1073/pnas.2401692121] [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: 01/25/2024] [Accepted: 10/02/2024] [Indexed: 11/20/2024] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most widely used rodent model for multiple sclerosis. Interferon-γ (IFN-γ) and regulatory T cells (Tregs) are individually well known to play beneficial roles in amelioration of EAE. However, little is known about the relationship between IFN-γ and Tregs during the disease. Here, we show that IFN-γ polarizes Tregs into T helper 1 (Th1)-type Tregs (Th1-Tregs) to recover from EAE. Single-cell RNA sequencing analysis revealed that brain Tregs showed signs of IFN-γ stimulation during EAE. Loss of IFN-γ signaling in Tregs and of T cell-derived IFN-γ impaired the Th1-Treg polarization and worsened the disease. Moreover, selective ablation of Th1-Tregs using an intersectional genetic method promoted proinflammatory features of macrophages in the inflamed brains and exacerbated the EAE. Taken together, our study highlights a critical role of T cell-derived IFN-γ for Th1-Treg polarization in inflamed brain to ameliorate EAE.
Collapse
Affiliation(s)
- Masaaki Okamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Ayumi Kuratani
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita879-5593, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita879-5593, Japan
- Division of Pathophysiology, Research Center for GLOBAL and LOCAL Infectious Diseases, Oita University, Oita879-5593, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka565-0871, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka565-0871, Japan
| |
Collapse
|
43
|
Zhong X, Wu H, Zhang W, Shi Y, Gwack Y, Xue HH, Sun Z. Distinct RORγt-dependent Th17 immune responses are required for autoimmune pathogenesis and protection against bacterial infection. Cell Rep 2024; 43:114951. [PMID: 39504243 PMCID: PMC11931457 DOI: 10.1016/j.celrep.2024.114951] [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: 05/02/2024] [Revised: 09/23/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
T helper (Th)17 cells mediate both protective anti-bacterial immune responses and autoimmune pathogenesis, but the distinct pathways regulating these Th17 responses remain unclear. Retinoid-related orphan receptor γ t (RORγt) is a master transcription factor that governs Th17 cell generation and effector functions. We found that a K256R mutation in RORγt impairs Th17-mediated experimental autoimmune encephalomyelitis (EAE) without affecting the clearance of Citrobacter rodentium. This indicates distinct RORγt roles in central nervous system pathogenesis versus gut-associated protective Th17 responses. Mechanically, RORγt/Runx1-dependent upregulation of galectin-3 (Lgals3) and chemokine receptor Ccr6 in CD4+ T cells is essential for EAE development but not for bacterial clearance. Moreover, Lgals3 is selectively required for recruiting macrophages to produce interleukin (IL)-1β, which in turn promotes Ccr6 expression on CD4+ T cells during EAE pathogenesis. Our findings highlight different RORγt-regulated Th17 pathways in autoimmunity and anti-bacterial immunity, with implications for therapies targeting Th17-mediated autoimmunity while preserving effective anti-bacterial responses.
Collapse
Affiliation(s)
- Xiancai Zhong
- Department of Immunology & Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Hongmin Wu
- Department of Immunology & Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Wencan Zhang
- Department of Immunology & Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Yun Shi
- Department of Immunology & Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Hai-Hui Xue
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA
| | - Zuoming Sun
- Department of Immunology & Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
44
|
Wang H, Zhang Q, Sun Y, Tan W, Zhang M. AdipoR1 promotes pathogenic Th17 differentiation by regulating mitochondrial function through FUNDC1. J Biomed Res 2024; 38:1-12. [PMID: 39506876 DOI: 10.7555/jbr.38.20240244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Adiponectin receptor 1 ( Adipor1) deficiency has been shown to inhibit Th17 cell differentiation and reduce joint inflammation and bone erosion in antigen-induced arthritis (AIA) mice. Additional emerging evidence indicates that Th17 cells may differentiate into pathogenic (pTh17) and non-pathogenic (npTh17) cells, with the pTh17 cells playing a crucial role in numerous autoimmune and inflammatory conditions. In the current study, we found that Adipor1 deficiency inhibited pTh17 differentiation in vitro and that the deletion of Adipor1 in pTh17 cells reduced the mitochondrial function. RNA-sequencing (RNA-seq) demonstrated a significant increase in the expression levels of Fundc1, a gene related to mitochondrial function, in Adipor1-deficient CD4 + T cells. Interference with the Fundc1 expression in Adipor1-deficient CD4 + T cells partially mitigated the effect of Adipor1 deficiency on mitochondrial function and pTh17 differentiation. In conclusion, the current study demonstrated a novel role of AdipoR1 in regulating mitochondrial function via FUNDC1 to promote pTh17 cell differentiation, providing some insights into potential therapeutic targets for autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Hui Wang
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Qian Zhang
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yuankai Sun
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Wenfeng Tan
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Miaojia Zhang
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
45
|
Cuevas-Martínez R, González-Chávez SA, Bermúdez M, Salas-Leiva JS, Vázquez-Olvera G, Hinojos-Gallardo LC, Chaparro-Barrera E, Pacheco-Silva C, Romero-Sánchez C, Villegas-Mercado CE, Pacheco-Tena C. Intermittent fasting reduces inflammation and joint damage in a murine model of rheumatoid arthritis: insights from transcriptomic and metagenomic analyses. BMC Rheumatol 2024; 8:64. [PMID: 39587696 PMCID: PMC11587710 DOI: 10.1186/s41927-024-00436-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Intermittent fasting (IF) has shown benefits in various pathological conditions. Although its anti-inflammatory potential has been recognized, its effects on the mechanism underlying rheumatoid arthritis (RA) remain insufficiently characterized. This study aimed to investigate the effects of IF in a murine model of RA. METHODS Collagen-induced arthritis (CIA) was developed in sixteen male DBA/1 mice, randomly assigned to two groups, with one undergoing IF every other day for four weeks. The effects of IF on joint inflammation and remodeling were evaluated clinically, histologically, and through tomography. Transcriptomic changes were characterized using expression microarrays, validated by RT-qPCR, and confirmed by immunohistochemistry. Additionally, modifications in gut microbiota were assessed through 16 S sequencing. RESULTS Mice subjected to IF significantly reduced the incidence and severity of clinical arthritis. Histological and radiographic assessments confirmed a decrease in inflammation and joint damage. Transcriptomic analysis revealed that IF led to the upregulation of 364 genes and the downregulation of 543 genes, with notable reductions in inflammatory signaling pathways associated with RA-related genes, including Cd72, Cd79a, Ifna, Il33, and Bglap 2. Notably, IL33 emerged as a pivotal mediator in the inflammatory processes mitigated by fasting. Key regulators associated with IF effects, such as CEBPA, FOXO1, HIF1A, PPARG, and PPARA, were identified, indicating a complex interplay between metabolic and inflammatory pathways. Furthermore, differential expression of microRNAs and lncRNAs, including miR-15b, miR-103-2, miR-302a, miR-6985, and miR- 5624, was observed. Metagenomic analysis indicated that IF enhanced the abundance and diversity of the gut microbiome, explicitly promoting anti-inflammatory bacterial populations, notably within the genus Ruminococcaceae. CONCLUSION Our findings suggest that IF exerts significant anti-inflammatory and immunoregulatory effects in the context of CIA. Given its non-risky nature, further investigation into the potential benefits of IF in patients with RA is warranted. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Rubén Cuevas-Martínez
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico
| | - Susana Aideé González-Chávez
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico.
| | - Mercedes Bermúdez
- Faculty of Dentistry, Autonomous University of Chihuahua, Chihuahua, Mexico
| | | | | | | | - Eduardo Chaparro-Barrera
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico
| | - César Pacheco-Silva
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico
| | - Consuelo Romero-Sánchez
- Cellular and Molecular Immunology Group (INMUBO), School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | | | - César Pacheco-Tena
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico.
| |
Collapse
|
46
|
Yazdani R, Naziri H, Azizi G, Ciric B, Askari M, Ahmadi AM, Aseervatham J, Zhang GX, Rostami A. IL-37 suppresses CNS autoimmunity by increasing the frequency of Treg cells and reducing CD4 + T cell-derived IL-10 production. J Neuroinflammation 2024; 21:301. [PMID: 39563375 PMCID: PMC11575187 DOI: 10.1186/s12974-024-03295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Interleukin-37 (IL-37) has anti-inflammatory properties in innate and adaptive immunity. Patients with multiple sclerosis (MS), an autoimmune inflammatory demyelinating disease of the central nervous system (CNS), have increased serum levels of IL-37. However, it is unknown whether IL-37 has an inhibitory effect on ongoing autoimmune neuroinflammation, thus offering a potential MS therapy. AIM Here, we examined the effect of IL-37 in an experimental autoimmune encephalomyelitis (EAE) model after disease onset to determine if it was protective. FINDINGS IL-37-treated mice developed a less severe disease than control mice, with reduced demyelination as determined by increased expression of myelin basic protein. IL-37 suppressed inflammation by decreasing infiltration of CD4 + T cells into the CNS and increasing the frequency of regulatory T cells, while IL-10 expression by CD4 + T cells decreased over time in the CNS. CONCLUSION Our findings confirm the immunomodulatory role of IL-37 in CNS inflammation during ongoing disease, thus indicating the potential of IL-37 as an inhibitory reagent for MS therapy.
Collapse
Affiliation(s)
- Reza Yazdani
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Hamed Naziri
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Gholamreza Azizi
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Mozhde Askari
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Amir Moghadam Ahmadi
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Jaya Aseervatham
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA.
| |
Collapse
|
47
|
Becher B, Derfuss T, Liblau R. Targeting cytokine networks in neuroinflammatory diseases. Nat Rev Drug Discov 2024; 23:862-879. [PMID: 39261632 DOI: 10.1038/s41573-024-01026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
In neuroinflammatory diseases, systemic (blood-borne) leukocytes invade the central nervous system (CNS) and lead to tissue damage. A causal relationship between neuroinflammatory diseases and dysregulated cytokine networks is well established across several preclinical models. Cytokine dysregulation is also observed as an inadvertent effect of cancer immunotherapy, where it often leads to neuroinflammation. Neuroinflammatory diseases can be separated into those in which a pathogen is at the centre of the immune response and those of largely unknown aetiology. Here, we discuss the pathophysiology, cytokine networks and therapeutic landscape of 'sterile' neuroinflammatory diseases such as multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), neurosarcoidosis and immune effector cell-associated neurotoxicity syndrome (ICANS) triggered by cancer immunotherapy. Despite successes in targeting cytokine networks in preclinical models of neuroinflammation, the clinical translation of targeting cytokines and their receptors has shown mixed and often paradoxical responses.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Tobias Derfuss
- Department of Neurology and Biomedicine, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Roland Liblau
- Institute for inflammatory and infectious diseases, INSERM UMR1291 - CNRS UMR505, Toulouse, France.
| |
Collapse
|
48
|
Park JS, Song AY, Bae JY, Han JW, Kim TH, Kim CJ, Lee SK. IL-17 Producing T to Foxp3 +CD4 + Regulatory T Cell Ratio as a Diagnostic and Prognostic Marker in Women With Recurrent Pregnancy Loss and Its Implications for Intravenous Immunoglobulin Therapy. Am J Reprod Immunol 2024; 92:e70020. [PMID: 39584290 DOI: 10.1111/aji.70020] [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: 08/26/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024] Open
Abstract
PROBLEM The imbalance in the Th17/Regulatory T (Treg) cell ratio is associated with recurrent pregnancy loss (RPL). This study aimed to determine a cut-off for the Th17/Treg cell ratio to predict pregnancy outcomes in RPL and evaluate the effectiveness of intravenous immunoglobulin (IVIG) based on this cut-off value. METHOD OF STUDY This retrospective cohort study included 49 idiopathic RPL and 75 controls. The subgroups of IL-17+ T cell to Foxp3+ T cell ratios in peripheral blood were measured using flow cytometry. The cut-off values of Th17/Treg cell ratios were determined by the ROC curve to distinguish between RPL and controls. The IVIG treatment effectiveness in pregnancy outcome was compared between high- and low-ratio groups. Pearson correlation assessed the Th17/Treg cell ratio's relationship with NK cell cytotoxicity (NKC), NK cell percentage, and Th1/Th2 cell ratio. RESULTS Using the ROC curve, we identified six Th17/Treg cell ratio markers with diagnostic value, and the following two, CD3+IL-17+ T cell/CD3+Foxp3high T cell ratio (sensitivity at 97%) and CD4+IL-17+ T cell/CD3+Foxp3high T cell ratio (specificity at 93.61%), showed the highest statistical significance in diagnosing idiopathic RPL. Among the six diagnostic markers, in terms of predicting pregnancy outcomes with IVIG treatment, CD3+IL-17+ T cell/CD4+Foxp3+ T cell ratio was the most valuable prognostic marker. In RPL women with high CD3+IL-17+ T cell/CD4+Foxp3+ T cell ratio (≥ 1.096), the live birth rate (LBR) was improved with IVIG treatment. (IVIG treatment, 78.57% vs. no IVIG, 28.57%, p = 0.026). On the other hand, RPL women with low CD3+IL-17+ T cell/CD4+Foxp3+ T cell ratio did not demonstrate the effectiveness of IVIG (LBRs with IVIG treatment, 50.00% vs. no IVIG, 84.62%, p = 0.219). In a correlation study, the CD3+IL-17+ T cell/CD4+Foxp3+ T cell ratio was an independent prognostic marker, showing no correlation with NKC, NK cell percentage, and Th1/Th2 cell ratio. CONCLUSION The CD3+IL-17+ T/CD4+Foxp3+ T cell ratio may serve as a valuable marker for understanding the pathogenesis of RPL, predicting pregnancy outcomes, and selecting candidates for immunotherapy. Our study demonstrates that IVIG treatment can significantly improve LBR in women with a high CD3+IL-17+ T/CD4+Foxp3+ T ratio, offering a promising therapeutic approach for this challenging condition.
Collapse
Affiliation(s)
- Jin-Sol Park
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ah-Yun Song
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Republic of Korea
| | - Ju-Young Bae
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Republic of Korea
| | - Jae Won Han
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Republic of Korea
| | - Tae Hyun Kim
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Republic of Korea
| | - Chul-Jung Kim
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Republic of Korea
| | - Sung Ki Lee
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Republic of Korea
| |
Collapse
|
49
|
Masuyama S, Mizui M, Morita M, Shigeki T, Kato H, Yamamoto T, Sakaguchi Y, Inoue K, Namba-Hamano T, Matsui I, Okuno T, Yamamoto R, Takashima S, Isaka Y. Enhanced fatty acid oxidation by selective activation of PPARα alleviates autoimmunity through metabolic transformation in T-cells. Clin Immunol 2024; 268:110357. [PMID: 39243921 DOI: 10.1016/j.clim.2024.110357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
While fatty acid oxidation (FAO) in mitochondria is a primary energy source for quiescent lymphocytes, the impact of promoting FAO in activated lymphocytes undergoing metabolic reprogramming remains unclear. Here, we demonstrate that pemafibrate, a selective PPARα modulator used clinically for the treatment of hypertriglyceridemia, transforms metabolic system of T-cells and alleviates several autoimmune diseases. Pemafibrate suppresses Th17 cells but not Th1 cells, through the inhibition of glutaminolysis and glycolysis initiated by enhanced FAO. In contrast, a conventional PPARα agonist fenofibrate significantly inhibits cell growth by restraining overall metabolisms even at a dose insufficient to induce fatty acid oxidation. Clinically, patients receiving pemafibrate showed a significant decrease of Th17/Treg ratio in peripheral blood. Our results suggest that augmented FAO by pemafibrate-mediated selective activation of PPARα restrains metabolic programs of Th17 cells and could be a viable option for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Satoshi Masuyama
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Masashi Morita
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; Department of Nephrology, NHO Osaka Minami Medical Center, Japan
| | - Takatomo Shigeki
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yusuke Sakaguchi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazunori Inoue
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomoko Namba-Hamano
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryohei Yamamoto
- Department of Health Promotion Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
50
|
Tiligada E, Stefanaki C, Ennis M, Neumann D. Opportunities and challenges in the therapeutic exploitation of histamine and histamine receptor pharmacology in inflammation-driven disorders. Pharmacol Ther 2024; 263:108722. [PMID: 39306197 DOI: 10.1016/j.pharmthera.2024.108722] [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: 05/24/2024] [Revised: 07/31/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Inflammation-driven diseases encompass a wide array of pathological conditions characterised by immune system dysregulation leading to tissue damage and dysfunction. Among the myriad of mediators involved in the regulation of inflammation, histamine has emerged as a key modulatory player. Histamine elicits its actions through four rhodopsin-like G-protein-coupled receptors (GPCRs), named chronologically in order of discovery as histamine H1, H2, H3 and H4 receptors (H1-4R). The relatively low affinity H1R and H2R play pivotal roles in mediating allergic inflammation and gastric acid secretion, respectively, whereas the high affinity H3R and H4R are primarily linked to neurotransmission and immunomodulation, respectively. Importantly, however, besides the H4R, both H1R and H2R are also crucial in driving immune responses, the H2R tending to promote yet ill-defined and unexploited suppressive, protective and/or resolving processes. The modulatory action of histamine via its receptors on inflammatory cells is described in detail. The potential therapeutic value of the most recently discovered H4R in inflammatory disorders is illustrated via a selection of preclinical models. The clinical trials with antagonists of this receptor are discussed and possible reasons for their lack of success described.
Collapse
Affiliation(s)
- Ekaterini Tiligada
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Charikleia Stefanaki
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; University Research Institute of Maternal and Child Health and Precision Medicine, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Madeleine Ennis
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Detlef Neumann
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| |
Collapse
|