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
|
Spunde K, Korotkaja K, Sominskaya I, Zajakina A. Genetic adjuvants: A paradigm shift in vaccine development and immune modulation. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102536. [PMID: 40336572 PMCID: PMC12056970 DOI: 10.1016/j.omtn.2025.102536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
The COVID-19 pandemic underscored the urgency of developing effective vaccines to combat infectious diseases, especially in vulnerable populations such as the elderly and immunocompromised. While recombinant protein vaccines offer safety, their poor immunogenicity highlights the need for advanced vaccination platforms. New genetic/nucleic acid vaccine formulations like plasmid DNA and mRNA showed efficiency and safety in preclinical and clinical studies; however, they demand innovative adjuvants because their mechanism of action differs from traditional protein vaccines. Genetic adjuvants-encoded by nucleic acids within DNA, RNA, or viral vectors-emerge as a promising solution by targeting and modulating specific immune pathways, including antigen presentation, T cell activation, and memory formation. These innovative adjuvants enhance vaccine efficacy by fine-tuning innate and adaptive immune responses, overcoming immune senescence, and addressing the challenges of CD8+ T cell activation in immunocompromised populations. This review explores the potential of genetically encoded adjuvants, including cytokines, chemokines, and other immune modulators. By comparing these adjuvants to traditional formulations, we highlight their capacity to address the limitations of modern vaccines while discussing their integration with emerging technologies like RNA-based vaccines. As genetic adjuvants advance toward clinical application, understanding their mechanisms and optimizing their delivery is pivotal to unlocking next-generation immunization strategies.
Collapse
Affiliation(s)
- Karina Spunde
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Ksenija Korotkaja
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Irina Sominskaya
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Anna Zajakina
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| |
Collapse
|
3
|
Senapati S, Bertolini TB, Minnier MA, Yazicioglu MN, Markusic DM, Zhang R, Wicks J, Nahvi A, Herzog RW, Walsh MC, Cejas PJ, Armour SM. Inhibition of IFNAR-JAK signaling enhances tolerability and transgene expression of systemic non-viral DNA delivery. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102502. [PMID: 40206655 PMCID: PMC11979999 DOI: 10.1016/j.omtn.2025.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Lipid nanoparticles (LNPs) have demonstrated significant therapeutic value for non-viral delivery of mRNA and siRNA. While there is considerable interest in utilizing LNPs for delivering DNA (DNA-LNPs) to address a broad range of genetic disorders, acute inflammatory responses pose significant safety concerns and limit transgene expression below therapeutically relevant levels. However, the mechanisms and immune signaling pathways underlying DNA-LNP-triggered inflammatory responses are not well characterized. Through the use of gene-targeted mouse models, we have identified cGAS-STING and interferon-α/β receptor (IFNAR) pathways as major mediators of acute inflammation triggered by systemic delivery of DNA-LNPs. cGAS-STING activation induces expression of numerous JAK-STAT-activating cytokines, and we show that treatment of mice with the JAK inhibitors ruxolitinib or baricitinib significantly improves tolerability to systemically delivered DNA-LNPs. Furthermore, specific inhibition of IFNAR signaling enhances both DNA-LNP tolerability and transgene expression. Utilization of JAK inhibitors or IFNAR blockade represent promising strategies for enhancing the safety and efficacy of non-viral DNA delivery for gene therapy.
Collapse
Affiliation(s)
| | - Thais B. Bertolini
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - David M. Markusic
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rui Zhang
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Joan Wicks
- Gene Therapy Research, Spark Therapeutics, Philadelphia, PA, USA
| | - Ali Nahvi
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Roland W. Herzog
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Pedro J. Cejas
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Sean M. Armour
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| |
Collapse
|
4
|
Si H, Cui Y, Zu J, Shi Y, Song Y, Zhen Y, Li S. IL-18 favors Th2 responses in sporotrichosis caused by Sporothrix globosa, prolonging the course of the disease. PLoS Negl Trop Dis 2025; 19:e0013170. [PMID: 40489556 DOI: 10.1371/journal.pntd.0013170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Accepted: 05/22/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Sporotrichosis is a subcutaneous mycosis caused by members of the genus Sporothrix, among which Sporothrix globosa (S. globosa) is the predominant etiological agent in Asia. T cell immunity plays crucial roles in fungal infections, yet the local T cell immune profile in sporotrichosis lesions remains unclear. IL-18, a pleiotropic cytokine capable of modulating T cell responses, is also poorly understood in host defense against S. globosa. METHODOLOGY/PRINCIPAL FINDINGS qPCR, western blot, and IHC/mIHC were employed to profile IL-18, IL-18 BP, caspase-1 and IL-18R axis, along with Th1,Th2, and Th17 cells and their specific cytokines in sporotrichosis lesions versus healthy skin. Cellular sources of IL-18 in the lesions were identified via mIHC, and IL-18 production from keratinocyte/PBMCs exposed to S. globosa in vitro was tested by qPCR and western blot. Flow cytometry was performed to evaluate the role of IL-18 in Th2 polarization in the PBMCs-S. globosa coculture system. Lesional skin exhibited hyperactivated IL-18 signaling, marked by upregulated IL-18, caspase-1, and IL-18R, alongside decreased IL-18 BP. IL-18 was primarily released by dermal dendritic cells and Langerhans cells, rather than keratinocytes. A mixed Th1/Th2/Th17 microenvironment with Th2 predominance correlated strongly with prolonged disease duration. Strikingly, IL-18 synergistically interacted with Th1-derived IL-2 to establish a self-reinforcing Th2 loop, as evidenced by the strong correlation between lesional IL-4 and IL-18 levels (r = 0.70) and the concomitant upregulation of IL-18/IL-2 during S. globosa-induced Th2 expansion in PBMCs-an effect reversed by IL-18/IL-2 neutralizing antibodies. Critically, this Th2 skewing was mechanistically dependent on NF-κB signaling, as demonstrated through pharmacological pathway inhibition. CONCLUSIONS/SIGNIFICANCE This study unveils the dual role of IL-18 in human sporotrichosis caused by S. globosa-amplifying both Th1 and Th2 responses but ultimately driving pathogenic Th2 polarization through IL-2 crosstalk. Our work identifies IL-18/IL-2/NF-κB axis as a key Th2-polarizing mechanism driving chronicity in this disease. Targeting this axis could recalibrate anti-fungal immunity, offering translational strategies for this subcutaneous disease.
Collapse
Affiliation(s)
- Henan Si
- Department of Dermatology and Venerology, the First Hospital of Jilin University, Changchun, China
| | - Yan Cui
- Department of Dermatology and Venerology, the First Hospital of Jilin University, Changchun, China
| | - Jianjiao Zu
- Department of Dermatology and Venerology, the First Hospital of Jilin University, Changchun, China
| | - Ying Shi
- Department of Dermatology and Venerology, the First Hospital of Jilin University, Changchun, China
| | - Yang Song
- Department of Dermatology and Venerology, the First Hospital of Jilin University, Changchun, China
| | - Yu Zhen
- Department of Dermatology and Venerology, the First Hospital of Jilin University, Changchun, China
| | - Shanshan Li
- Department of Dermatology and Venerology, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Su X, Sun Y, Dai A. New insights into pulmonary arterial hypertension: interaction between PANoptosis and perivascular inflammatory responses. Apoptosis 2025; 30:1097-1116. [PMID: 39979525 DOI: 10.1007/s10495-025-02086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a heterogeneous disease characterized by various etiologies, with pulmonary vascular remodeling recognized as a main pathological change. Currently, it is widely accepted that vascular remodeling is closely associated with abnormal pulmonary vascular cell death and perivascular inflammation. The simultaneous activation of various pulmonary vascular cell death leads to immune cell adhesion and inflammatory mediator releases; And in turn, the inflammatory response may also trigger cell death and jointly promote the progression of vascular remodeling. Recently, PANoptosis has been identified as a phenomenon that describes the simultaneous activation and interaction of multiple forms of programmed cell death (PCD). Therefore, the relationship between PANoptosis and inflammation in PAH warrants further investigation. This review examines the mechanisms underlying apoptosis, necroptosis, pyroptosis, and inflammatory responses in PAH, with a focus on PANoptosis and its interactions with inflammation. And it aims to elucidate the significance of this emerging form of cell death and inflammation in the pathophysiology of PAH and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xianli Su
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Yinhui Sun
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
6
|
Wang M, Wang Z, Zhang G, Fan J. Interleukin-enhanced CAR-engineered immune cells in tumor immunotherapy: current insights and future perspectives. Cytokine 2025; 192:156973. [PMID: 40449036 DOI: 10.1016/j.cyto.2025.156973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/15/2025] [Accepted: 05/27/2025] [Indexed: 06/02/2025]
Abstract
Despite the remarkable clinical success of chimeric antigen receptor (CAR)-T cell therapy in hematologic malignancies, the therapeutic efficacy of conventional second-generation CAR-T cells in treating solid tumors remains suboptimal, primarily due to three major biological barriers: (1) the immunosuppressive tumor microenvironment (TME), (2) inadequate tumor infiltration capacity, and (3) T cell exhaustion mechanisms. To overcome these limitations, innovative fourth-generation "armored" CAR-T cell platforms have been engineered with integrated cytokine-secreting modules designed to potentiate anti-tumor responses through localized immunomodulation. These advanced cellular therapeutics achieve targeted delivery of various immunostimulatory cytokines directly within the TME, thereby orchestrating three critical therapeutic effects: (I) remodeling of the immunosuppressive niche, (II) enhancement of immune cell persistence, and (III) neutralization of immunosuppressive signaling networks. This comprehensive review systematically examines the translational applications of cytokine-secreting CAR-engineered immune cells, including CAR-T, CAR-NK, and CAR-iNKT cell platforms, in solid tumor immunotherapy, with particular emphasis on multiple classes of immunomodulatory cytokines that enhance cytotoxic potential, promote immune cell survival, and counteract TME-mediated immunosuppression. We critically evaluate preclinical and clinical evidence demonstrating the therapeutic efficacy of cytokine-armed CAR-engineered cells across various tumor models, including hematological malignancies, glioblastoma, and neuroblastoma. Furthermore, this review addresses current translational challenges, particularly cytokine-associated toxicity profiles and innovative strategies for achieving spatiotemporal control of cytokine release, while discussing their potential implications for advancing clinical outcomes in solid tumor immunotherapy.
Collapse
Affiliation(s)
- Min Wang
- Department of Neurology, The Second Hospital of Jilin University, Changchun 130022, China
| | - Zixuan Wang
- Beijing Institute of Biological Products Co., Ltd, Beijing 101149, China
| | - Guangji Zhang
- Chinese Institutes for Medical Research, Beijing, China.
| | - Jia Fan
- Department of Neurology, The Second Hospital of Jilin University, Changchun 130022, China.
| |
Collapse
|
7
|
Sancho-Saldaña A, Gil-Sánchez A, Quirant-Sánchez B, Boigues M, Canudes M, Peralta S, Solana MJ, González-Mingot C, Quibus L, Martínez-Cáceres E, Torres P, Hervás JV, Moreno-Magallon J, Brieva L. Profile of Cytokines Associated with SARS-CoV2 Seropositivity in Multiple Sclerosis Patients and Its Persistence over Six Months. J Clin Med 2025; 14:3736. [PMID: 40507498 PMCID: PMC12155705 DOI: 10.3390/jcm14113736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/08/2025] [Accepted: 05/18/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Patients with multiple sclerosis (pwMS) receiving disease-modifying therapies (DMTs) may exhibit altered immune responses to infections such as SARS-CoV-2. This study aimed to characterize the cytokine profiles associated with prior SARS-CoV-2 infection and to identify immune markers related to the persistence of the humoral response in pwMS. Methods: A total of 90 pwMS were recruited before the introduction of COVID-19 vaccination in Spain; 46 were seropositive-defined by the presence of IgG, IgM, or IgA antibodies against SARS-CoV-2-and 44 were seronegative. We compared baseline cytokine levels between groups and followed seropositive individuals for six months to assess IgG antibody persistence. Results: Seropositive patients showed significantly lower baseline levels of IL-10, IL-23, and IFN-α compared to seronegative individuals. Notably, elevated IL-18 at baseline was associated with persistent IgG seropositivity at six months. Conclusions: These findings suggest a distinct cytokine profile in SARS-CoV-2-exposed pwMS and highlight IL-18 as a potential marker of sustained humoral response. This study provides insight into host-virus immune dynamics in MS patients and may help guide future strategies for infection monitoring and immune evaluation in this population.
Collapse
Affiliation(s)
- Agustín Sancho-Saldaña
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Anna Gil-Sánchez
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Bibiana Quirant-Sánchez
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
- Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Cerdanyola del Valles, Spain
| | - Marc Boigues
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
| | - Marc Canudes
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Silvia Peralta
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - María José Solana
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Cristina González-Mingot
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Department of Medicine, University of Lleida (UdL), 25198 Lleida, Spain
| | - Laura Quibus
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Eva Martínez-Cáceres
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
- Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Cerdanyola del Valles, Spain
| | - Pascual Torres
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - José Vicente Hervás
- Servicio de Neurología, Hospital de Sant Joan Despí Moisés Broggi, 08970 Barcelona, Spain;
| | - Judith Moreno-Magallon
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Luis Brieva
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Department of Medicine, University of Lleida (UdL), 25198 Lleida, Spain
| |
Collapse
|
8
|
Kim HK, Ju H, Chung YH, Ju ES, Cho Y, Jeon Y, Jo DG, Min JH. Serum sEV miRNAs as Biomarkers in Myelin Oligodendrocyte Glycoprotein Antibody-Associated Disease. Mol Neurobiol 2025:10.1007/s12035-025-04932-3. [PMID: 40388105 DOI: 10.1007/s12035-025-04932-3] [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: 09/06/2024] [Accepted: 04/07/2025] [Indexed: 05/20/2025]
Abstract
Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) is a distinct CNS demyelinating disorder that differs from multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). However, diagnosing MOGAD remains challenging due to the need to clinically exclude similar conditions and the variability in assay results. While liquid biomarkers have been extensively studied in MS and NMOSD, research on biomarkers for MOGAD remains limited. This study aims to investigate serum-derived small extracellular vesicle (sEV) miRNAs as potential diagnostic and prognostic biomarkers for MOGAD, distinguishing it from MS, NMOSD, and healthy controls. A comprehensive analysis of miRNAs in serum-derived sEVs was conducted to identify differentially expressed miRNAs among the groups. Correlations between miRNA profiles and clinical parameters, including the expanded disability status scale (EDSS) score and annualized relapse rate (ARR), were examined. The diagnostic potential of miRNAs was evaluated using the area under the curve (AUC) in the receiver operating characteristics (ROC) analyses. Serum samples were obtained from 47 patients (N = 11, MOGAD; N = 12, MS; and N = 12, NMOSD) and 12 healthy controls (HCs). We identified 77 dysregulated miRNAs in MOGAD patients, compared to HCs. Each three-miRNA panel demonstrated the highest AUC values for distinguishing MOGAD from HC (1.000), MOGAD from MS (0.939), and MOGAD from NMOSD (1.000). Additionally, hsa-miR-924 exhibited the strongest correlation with the EDSS score (ρ = -0.67, p < 0.001), while hsa-miR-548i showed the strongest correlation with ARR (ρ = -0.69, p < 0.001) in MOGAD. These miRNAs are involved in various pathways, including neuronal development, immune response, synaptic function, and chromatin remodeling, highlighting their potential roles in the pathophysiology of MOGAD. Serum sEV-derived miRNAs show strong potential as biomarkers for MOGAD, offering high diagnostic accuracy and correlations with clinical parameters. These findings pave the way for improved diagnostic and therapeutic strategies in MOGAD; however, further validation in larger cohorts is necessary.
Collapse
Affiliation(s)
- Hark Kyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyunjin Ju
- Department of Neurology, Soonchunhyang University Seoul Hospital, Seoul, 04401, Republic of Korea
- Department of Neurology, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Yeon Hak Chung
- Department of Neurology, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
- Department of Neurology, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Eun-Seon Ju
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Yongeun Cho
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yeji Jeon
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Ju-Hong Min
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea.
- Department of Neurology, Samsung Medical Center, Seoul, 06351, Republic of Korea.
- Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, 06351, Republic of Korea.
| |
Collapse
|
9
|
Zhang Y, Guan Y, Dai M, Yang Y, Yang F. Microcystin-LR induces lung injury in mice through the NF-κB/NLRP3 pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:385-394. [PMID: 39773316 DOI: 10.1080/15287394.2024.2443525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Microcystin-LR (MC-LR) a cyclic toxin produced by cyanobacterial species is known to exert detrimental effects on various organs, including lung. Several investigators demonstrated that MC-LR exerts pulmonary toxicity, but the underlying mechanisms remain unclear. This study aimed to investigate whether exposure to MC-LR-induced lung inflammation and examine the underlying mechanisms. Thirty specific pathogen-free (SPF) male mice were allocated into control and MC-LR treatment groups. Mice were intraperitoneally injected with physiological saline or MC-LR (20 μg/kg) daily for a total of 21 days. Our findings indicated that exposure to MC-LR-produced histopathological changes in lung tissue, including thickening of alveolar walls and inflammatory infiltration. MC-LR was found to upregulate mRNA expression levels of pro-inflammatory cytokines TNFα, IL-6, IL-1β, and IL-18. Further, MC-LR significantly elevated the expression levels of proteins associated with the NF-κB/NLRP3 pathway p-NF-κB, NLRP3, Caspase-1, ASC. The activation of NF-κB/NLRP3 pathway further promoted the release of inflammatory cytokine IL-1β and cleavage of pyroptosis-associated GSDMD protein. These findings indicate that MC-LR may induce lung inflammation by promoting cell pyroptosis via the activation of the NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Yin Zhang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ying Guan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Manni Dai
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yue Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| |
Collapse
|
10
|
Vandenberg EG, Kelly SB, Zahra VA, Lu H, Thiel A, Hooper SB, Galinsky R, Polglase GR. Investigating pulmonary inflammation and injury after progressive systemic inflammation in preterm fetal sheep. Front Physiol 2025; 16:1542613. [PMID: 40443447 PMCID: PMC12120474 DOI: 10.3389/fphys.2025.1542613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Preterm birth and intrauterine inflammation are commonly associated with lung inflammation and remodeling. We developed a fetal inflammatory response model using increasing doses of intravenous lipopolysaccharide (LPS) to cause systemic inflammation and injury. However, the effects of an increasing systemic inflammatory response on fetal lung inflammation and injury are not known. We aimed to investigate the effect of repeated increasing doses of intravenous LPS on pulmonary inflammation and injury in preterm fetal sheep. Methods Fetal sheep at 124 days of gestation (term ∼148 days) underwent surgical instrumentation. At 129 days of gestation, fetal sheep were randomized to saline control (n = 8) or repeated LPS infusions (300 ng/24 h then doubled every 24 h for 2 days; n = 8). Four days after LPS/saline infusions commenced, fetal lungs were collected for histological and molecular analysis of markers of pulmonary inflammation and injury. Results Repeated increasing doses of intravenous LPS decreased arterial pH, PaO2, SaO2 and increased lactate and PaCO2 compared to controls. LPS infusions caused a decrease in mRNA expression of pro-inflammatory cytokines IL1B (p = 0.030) and IL6 (p = 0.034) and an increase in IL18 (p < 0.0001). LPS exposure did not alter histological assessment of airway structure, elastin or collagen abundance, inflammatory cell infiltration or cell death compared to controls. Conclusion Intravenous administration of LPS did not cause fetal lung inflammation and injury assessed 4 days after LPS infusions commenced. Direct exposure to endotoxins within the lungs may be necessary to induce inflammation and injury in the fetal lungs.
Collapse
Affiliation(s)
- E. G. Vandenberg
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - S. B. Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - V. A. Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - H. Lu
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - A. Thiel
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - S. B. Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - R. Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - G. R. Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
11
|
Zańko A, Pawłowski M, Milewski R. The Impact of Physical Exercise on Male Fertility Through Its Association with Various Processes and Aspects of Human Biology. J Clin Med 2025; 14:3442. [PMID: 40429435 PMCID: PMC12112722 DOI: 10.3390/jcm14103442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objective: Infertility affects approximately 10-15% couples in industrialized countries. It has numerous causes, including genetic and environmental factors, lifestyle choices, and physiological disorders. The increasing prevalence of infertility underlines the importance of research into interventions to improve reproductive health, with a strong focus on physical activity. Infertility research was traditionally mainly directed toward female health. Although the male factor is being increasingly accepted as being equally important, this area remains under-researched. The current review focuses on the impact of physical activity on male fertility through its effects on immune function, the cardiovascular system, hormonal balance, metabolism, and physical interaction with the male reproductive system. Materials and Methods: A comprehensive literature review of studies addressing the effects of physical activity on male fertility was conducted using PubMed/Medline, Scopus, and the Web of Science. Mostly recent studies were included, with a small number of older ones included in cases when their content remains relevant. The review focused on articles studying the processes involved and associations between physical activity and male fertility through immune and cardiovascular effects, endocrine modulation, the influence on obesity and insulin metabolism, and the physical impact on the body. Results: The findings revealed the existence of a fairly strong consensus that moderate physical activity enhances semen quality, hormonal balance, and metabolic health, positively influencing male fertility. Physical activity reduces inflammation and oxidative stress, enhances cardiovascular functioning, and contributes to oxygen and nutrient supply to the reproductive organs. On the contrary, strenuous training can adversely affect fertility, mostly through hormonal disruption and oxidative stress. It can also have various indirect effects on fertility through sports-related behavior and incidents, such as wearing tight-fitting clothes or overheating. Conclusions: Physical activity can affect male fertility in numerous ways, positively influencing reproductive health when performed at a moderate intensity. Understanding the balance between beneficial and excessive exercise as well as the impact of incidental factors related to performing sports regularly are, thus, extremely important in optimizing lifestyle-oriented interventions aimed at male fertility improvement.
Collapse
Affiliation(s)
| | | | - Robert Milewski
- Department of Biostatistics and Medical Informatics, Medical University of Białystok, 15-295 Białystok, Poland; (A.Z.); (M.P.)
| |
Collapse
|
12
|
Singh DD. NLRP3 inflammasome: structure, mechanism, drug-induced organ toxicity, therapeutic strategies, and future perspectives. RSC Med Chem 2025:d5md00167f. [PMID: 40370650 PMCID: PMC12070810 DOI: 10.1039/d5md00167f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025] Open
Abstract
Drug-induced toxicity is an important issue in clinical medicine, which typically results in organ dysfunction and adverse health consequences. The family of NOD-like receptors (NLRs) includes intracellular proteins involved in recognizing pathogens and triggering innate immune responses, including the activation of the NLRP3 inflammasome. The NLRP3 (nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3) inflammasome is a critical component for both innate and adaptive immune responses and has been implicated in various drug-induced toxicities, including hepatic, renal, and cardiovascular diseases. The unusual activation of the NLRP3 inflammasome causes the release of pro-inflammatory cytokines, such as IL-1β and IL-18, which can lead to more damage to tissues. Targeting NLRP3 inflammasome is a potential therapeutic endeavour for suppressing drug-induced toxicity. This review provides insights into the mechanism, drug-induced organ toxicity, therapeutic strategies, and prospective therapeutic approaches of the NLRP3 inflammasome and summarizes the developing therapies that target the inflammasome unit. This review has taken up one of the foremost endeavours in understanding and inhibiting the NLRP3 inflammasome as a means of generating safer pharmacological therapies.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan Jaipur 303002 India +91 9450078260
| |
Collapse
|
13
|
Shushtari A, Ashayeri H, Salmannezhad A, Seyedmirzaei H, Rezaei N. Pro-inflammatory cytokines in myasthenia gravis: a systematic review and meta-analysis. Neurol Sci 2025:10.1007/s10072-025-08218-3. [PMID: 40347402 DOI: 10.1007/s10072-025-08218-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/27/2025] [Indexed: 05/12/2025]
Abstract
BACKGROUND Myasthenia gravis (MG) is an autoimmune neuromuscular disorder impacting muscle endplate components. Pro-inflammatory cytokines, particularly, might play pivotal roles in MG pathogenesis, influencing regulatory T cells and contributing to chronic inflammation. We did this systematic review and meta-analysis to address the conflicting results about pro-inflammatory cytokine profiles in MG. METHODS A thorough search was conducted in PubMed, Scopus, and Embase to find studies measuring interleukin (IL)-1 family (IL-1β, IL-18, IL-33, IL-36, IL-37), IL-6, and tumor necrosis factor-alpha (TNF-α) levels in MG patients' serum and controls. Selection criteria encompassed various MG types, including ocular and generalized, with and without thymoma, and acetylcholine receptor (AChR) antibody-positive and negative. RESULTS Of the 1843 identified studies, 16 met the inclusion criteria. The meta-analysis revealed a significant increase in serum TNF-α, IL-1β, and IL-33 level in MG patients compared to controls. The included studies also implied elevated levels of IL-18 in people with MG compared to controls and elevated levels of IL-18 and IL-33 in generalized MG compared to ocular MG. CONCLUSION Our study highlights the altered profiles of pro-inflammatory cytokines in MG.
Collapse
Affiliation(s)
- Ali Shushtari
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamidreza Ashayeri
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A JBI Centre of Excellence, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Salmannezhad
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Homa Seyedmirzaei
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Occupational Sleep Research Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Children's Medical Center Hospital, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran.
| |
Collapse
|
14
|
Sun H, Yao X, Jiao Y, Kong X, Han Y, Li Y, Ge J, Cao Y, Lu H, Wang P, Xu Y, Li J, Ding K, Gao X. DNA remnants in red blood cells enable early detection of cancer. Cell Res 2025:10.1038/s41422-025-01122-7. [PMID: 40341742 DOI: 10.1038/s41422-025-01122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
Cytoplasmic DNA emerges as a consequence of genomic instability. However, its potential role in disease diagnosis has yet to be fully explored. Here we analyzed DNA remnants in mature red blood cells (rbcDNA) from both healthy individuals and cancer patients. Our study unveiled distinct genomic profiles in rbcDNA from cancer patients with early-stage solid tumors compared to those of healthy donors. Significant changes in read counts at specific genomic regions within rbcDNA were identified in patients, which were termed tumor-associated rbcDNA features. These features demonstrated potential for highly accurate early-stage cancer detection, proposing a novel approach for cancer detection. Moreover, tumor-associated rbcDNA features were observed in tumor mouse models, with some features being conserved between mice and humans. Chronic, but not transient, up-regulation of interleukin-18 is essential for the development of these features by promoting DNA damage in bone marrow hematopoietic cells through the up-regulation of NR4A1. These results underscore the remote regulation of chromosomal stability in hematopoietic cells by solid tumors and propose tumor-associated rbcDNA features as a promising strategy for early cancer detection.
Collapse
Affiliation(s)
- Haobo Sun
- School of Basic Medical Science, Fudan University, Shanghai, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xingyun Yao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yurong Jiao
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiangxing Kong
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuehua Han
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jianping Ge
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Yanfei Cao
- Department of Gastroenterology, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hongsheng Lu
- Department of Pathology, Taizhou Central Hospital Taizhou University Hospital, Taizhou, Zhejiang, China
| | - Pingli Wang
- Department of Respiratory Medicine, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Xu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun Li
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofei Gao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Wang Y, Ye J, Luo Y, Weng Z, Lu Q, Xia C, Ma F, Kang F, Chen X, Zhang W. Molecular characterization and functional analysis of IL-18 in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2025; 163:110398. [PMID: 40347992 DOI: 10.1016/j.fsi.2025.110398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Interleukin-18 (IL-18), a pro-inflammatory cytokine of the IL-1 family, is crucial for protecting the host against pathogen infection in mammals. In this study, a IL-18 homolog gene was cloned from large yellow croaker (Larimichthys crocea) (LcIL-18), which has an open reading frame (ORF) of 609 bp that encodes a polypeptide of 202 amino acids. The LcIL-18 C-terminus contains a typical IL-1 family signature and a caspase cleavage site. Phylogenetic analysis showed that LcIL-18 was most closely related to IL-18 of Miichthys miiuy. It was found that LcIL-18 was constitutively expressed in all 12 tissues tested of large yellow croakers, with the highest expression in gills. The expression of LcIL-18 in head kidney, spleen, skin, gills, and liver showed a differential pattern following infection with Pseudomonas plecoglossicida and Vibrio alginolyticus. P. plecoglossicida strongly induced LcIL-18 expression in these tissues. Conversely, in the early stage of infection, V. alginolyticus significantly inhibited LcIL-18 expression in head kidney, spleen, skin, and gills, but not in the liver. In vitro, LPS, Poly(I:C), P. plecoglossicida, and V. alginolyticus significantly upregulated the expression of the LcIL-18 in large yellow croaker head kidney (LYCK) cells. Furthermore, recombinant LcIL-18 (rLcIL-18) significantly increased cell viability and upregulated the expression of pro-inflammatory cytokines (LcIL-1β, LcIL-6, and LcTNF-α1) in LYCK cells. Our findings therefore indicated that LcIL-18 was involved in pro-inflammatory response induced by pathogenic bacteria.
Collapse
Affiliation(s)
- Yongyang Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Jiajia Ye
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Yufen Luo
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Zufeng Weng
- Pingtan Comprehensive Experimental Zone Marine and Fisheries Law Enforcement Detachment, Pingtan, 350400, PR China
| | - Qi Lu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Changchang Xia
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Fanfan Ma
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Fuyu Kang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Xinhua Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China.
| | - Weini Zhang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China; University Key Lab for Integrated Chinese Traditional and Western Veterinary Medicine and Animal Healthcare in Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China.
| |
Collapse
|
16
|
Tyler R, Vizioli C, Barb J, Farokhnia M, Leggio L. Circulating Immune and Endocrine Markers in Currently Drinking and Abstinent Individuals With Alcohol Use Disorder and Controls. Addict Biol 2025; 30:e70039. [PMID: 40317574 PMCID: PMC12046569 DOI: 10.1111/adb.70039] [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: 02/20/2025] [Revised: 02/20/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Alcohol use disorder (AUD) is associated with changes in endocrine and immune system function. This study is a secondary analysis aimed at investigating changes in circulating immune and endocrine biomarkers in blood samples from three groups: (1) healthy controls (HC, N = 12), (2) AUD-currently drinking, nontreatment seeking (CD, N = 9), and (3) AUD-abstinent, treatment-seeking (AB, N = 10; abstinent for at least 6 weeks). We hypothesized that both immune and endocrine biomarker concentrations would be different in AUD groups compared to healthy controls. Immune biomarkers included IL-8, IL-18, CCL2, TNF-α, IL-1RA, IL-6, and IL-10. Endocrine biomarkers included brain-derived neurotrophic factor (BDNF), glucagon-like peptide 1 (GLP-1), ghrelin, gastric inhibitory peptide (GIP), growth hormone, leptin, and insulin. Biomarker concentrations were compared between the three groups while controlling for age and sex, and associations between biomarker concentrations and behavioral measures were explored. IL-8 concentrations were elevated in AB compared to CD and HC (F(2,29) = 6.33, p = 0.006, ƞp 2 = 0.318). BDNF concentrations were lower in AB compared to HC (F(2,30) = 4.34, p = 0.02, ƞp 2 = 0.266). GLP-1 concentrations were higher in AB compared to HC (F(2,25) = 4.22, p = 0.03, ƞp 2 = 0.287). Exploratory analyses in combined groups showed that measures of past drinking, AUD severity, and anxiety/depression positively correlated with IL-18 and TNF-α and negatively correlated with BDNF. These results demonstrate that circulating concentrations of both immune and endocrine proteins are altered in abstinent individuals with a history of severe AUD (AB group) compared to healthy controls. In contrast, no group differences were observed for any biomarker between the nontreatment seeking, currently drinking people with AUD and the HC group. Our findings highlight the importance of accounting for AUD severity, comorbidities, and treatment-seeking status, especially when studying alcohol-related biomarkers.
Collapse
Affiliation(s)
- Ryan E. Tyler
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology (CPN) Section, Translational Addiction Medicine BranchNational Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of HealthBaltimoreMarylandUSA
- National Institute of General Medical SciencesBethesdaMarylandUSA
| | - Carlotta Vizioli
- Interoceptive Disorders Unit, Office of the Clinical DirectorNational Institute of Neurological Disorders and Stroke, NIHBethesdaMarylandUSA
| | - Jennifer J. Barb
- Translational Biobehavioral and Health Disparities Branch, Clinical Center, NIHBethesdaMarylandUSA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology (CPN) Section, Translational Addiction Medicine BranchNational Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of HealthBaltimoreMarylandUSA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology (CPN) Section, Translational Addiction Medicine BranchNational Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
17
|
Li M, Lou S, Chen K, Dong Y, Wang S, Yu T, Deng X, Li S. Oxidative stress induced by combined glyphosate and TBBPA exposure promotes gill autophagy and inflammation via the PI3K/AKT/mTOR pathway. FISH & SHELLFISH IMMUNOLOGY 2025; 160:110190. [PMID: 40020950 DOI: 10.1016/j.fsi.2025.110190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/25/2025] [Accepted: 02/10/2025] [Indexed: 03/03/2025]
Abstract
Glyphosate and tetrabromobisphenol A (TBBPA) are pollutants that pose a serious threat to the ecological safety of aquatic environments. However, there has been no report on the effects of combined exposure on the toxicity of carp fish gills in water. Therefore, we constructed a model of carp gill tissue and the carp epithelioma cells (EPC) cells exposed to glyphosate and/or TBBPA in vitro and in vivo, established a control group, a glyphosate group, a TBBPA group, and a glyphosate + TBBPA group, and added PI3K/AKT pathway activator musk ketone in vitro to verify the relationship between toxins and pathways. qRT-PCR and western blotting methods were used to detect the expression of oxidative stress-related indicators (CAT, GSH-Px, T-AOC, H2O2) and related genes. In vitro and in vivo results showed that glyphosate and/or TBBPA exposure resulted in overproduction of ROS, decreased activity of CAT, GSH-Px, T-AOC, and increased H2O2 content. Glyphosate and/or TBBPA exposure inhibited the PI3K/AKT/mTOR signaling pathway, further resulting in increased autophagy related genes LC3, ATG-5, Beclin-1, and decreased p62 expression. Inflammation related genes TNF-α, IL-1β, IL-6, IL-18 increased. And it was more significant when exposed in combination than when exposed alone. The addition of PI3K/AKT signaling pathway activator musk ketone in vitro can significantly alleviate the changes of autophagy and inflammation-related indicators. In summary, glyphosate and/or TBBPA induce oxidative stress by promoting gill autophagy and inflammation via the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Mingyue Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Shuang Lou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Kai Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yuting Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Shize Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tingting Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xinrui Deng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
18
|
Sardag I, Duvenci ZS, Belkaya S, Timucin E. Computational modeling of the anti-inflammatory complexes of IL37. J Mol Graph Model 2025; 136:108952. [PMID: 39854883 DOI: 10.1016/j.jmgm.2025.108952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Interleukin (IL) 37 is an anti-inflammatory cytokine belonging to the IL1 protein family. Owing to its pivotal role in modulating immune responses, elucidating the IL37 complex structures holds substantial therapeutic promise for various autoimmune disorders and cancers. However, none of the structures of IL37 complexes have been experimentally characterized. This computational study aims to address this gap through molecular modeling and classical molecular dynamics simulations. We modeled all protein-protein complexes of IL37 using a range of methods from homology modeling to AlphaFold2 multimer predictions. Models that successfully recapitulated experimental features underwent further analysis through molecular dynamics simulations. As positive controls, binary and ternary complexes of IL18 from PDB were included for comparison. Several key findings emerged from the comparative analysis of IL37 and IL18 complexes. IL37 complexes exhibited higher mobility than the IL18 complexes. Simulations of the IL37-IL18Rα complex revealed altered receptor conformations capable of accommodating a dimeric IL37, with the N-terminal loop of IL37 contributing significantly to complex mobility. Additionally, the glycosyl chain on N297 of IL18Rα, which contours one edge of the cytokine binding surface, acted as a steric block against the N-terminal loop of IL37. Further, investigations into interactions between IL37 and IL18BP suggested that a binding mode homologous to IL18 was unstable for IL37, indicating an alternative binding mechanism. Altogether, this study accesses to the structure and dynamics of IL37 complexes, revealing the structural underpinnings of the IL37's modulatory effect on the IL18 signaling pathway.
Collapse
Affiliation(s)
- Inci Sardag
- Bogazici University, Department of Molecular Biology and Genetics, Istanbul 34342, Turkey
| | - Zeynep Sevval Duvenci
- Acibadem University, Institute of Health Sciences Department of Biostatistics and Bioinformatics, Istanbul 34752, Turkey
| | - Serkan Belkaya
- Bilkent University, Department of Molecular Biology and Genetics, Ankara 06800, Turkey
| | - Emel Timucin
- Acibadem University, Institute of Health Sciences Department of Biostatistics and Bioinformatics, Istanbul 34752, Turkey; Acibadem University, School of Medicine Biostatistics and Medical Informatics, Istanbul 34752, Turkey.
| |
Collapse
|
19
|
Panday R, Rogy KM, Han YD, Khetani SR. Engineered microtissues to model the effects of dynamic heterotypic cell signaling on iPSC-derived human hepatocyte maturation. Acta Biomater 2025; 197:135-151. [PMID: 40089127 DOI: 10.1016/j.actbio.2025.03.020] [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: 08/14/2024] [Revised: 01/21/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
In vitro human liver models are indispensable for compound metabolism/toxicity screening, disease modeling, and regenerative medicine. While induced pluripotent stem cell-derived human hepatocyte-like cells (iHeps) mitigate the sourcing limitations with primary human hepatocytes (PHHs), their functional maturity is rate-limiting for application use. During development, immature hepatoblasts interact with different non-parenchymal cell (NPC) types, such as mesenchyme and endothelia, in a spatiotemporal manner to progress through functional maturation. Modeling such interactions in vitro is critical to elucidate the key regulators of iHep maturation. Here, we utilized high-throughput droplet microfluidics to encapsulate iHeps within monodisperse collagen I microgels (Ø ∼ 250 µm), which were coated with NPCs to generate 'microtissues' placed within microwells in multiwell plates. Embryonic fibroblasts and liver sinusoidal endothelial cells (LSECs) induced the highest level of iHep maturation over 4+ weeks of culture compared to adult hepatic stellate cells (myofibroblastic), liver portal fibroblasts, dermal fibroblasts, and human umbilical vein endothelial cells. Combining iHep microtissues in plates with Transwell inserts containing different NPC types enabled the modeling of dynamic heterotypic signaling on iHep maturation; introducing embryonic fibroblast signaling first, followed by LSECs, led to the highest iHep maturation. Unique cytokine secretion profiles were detected across the top-performing microtissue configurations; stromal-derived factor-1 alpha was validated as one factor that enhanced iHep maturation. Lastly, gene expression patterns and regulatory networks showed adult PHH-like maturation in LSEC/iHep microtissues compared to iHep-only microtissues. Overall, microtissues are useful for elucidating the microenvironmental determinants of iHep maturation and for future use in downstream applications. STATEMENT OF SIGNIFICANCE: Induced pluripotent stem cell-derived hepatocyte-like cells (iHeps) hold great promise for drug screening, disease modeling, and regenerative medicine but often exhibit immature phenotypes. We utilized high-throughput droplet microfluidics to generate 3D microtissues containing iHeps and non-parenchymal cell (NPC) types to elucidate the effects of dynamic NPC signaling on iHep maturation. We observed that iHep maturation is significantly enhanced with embryonic fibroblasts and liver sinusoidal endothelial cells (LSEC) compared to adult liver fibroblasts and non-liver endothelia; the LSEC/iHep microtissues showed adult liver-like gene expression signatures. The highest iHep maturation in microtissues was achieved when mesenchymal stimulation was introduced first, followed by LSEC stimulation. Our platform provides a robust framework to elucidate cellular and molecular mediators of iHep maturation and biomedical applications.
Collapse
Affiliation(s)
- Regeant Panday
- Department of Biomedical Engineering, University of Illinois Chicago, 851 S Morgan St, 218 SEO, Chicago, IL 60607, USA
| | - Kerry M Rogy
- Department of Biomedical Engineering, University of Illinois Chicago, 851 S Morgan St, 218 SEO, Chicago, IL 60607, USA
| | - Yong Duk Han
- Department of Biomedical Engineering, University of Illinois Chicago, 851 S Morgan St, 218 SEO, Chicago, IL 60607, USA
| | - Salman R Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, 851 S Morgan St, 218 SEO, Chicago, IL 60607, USA.
| |
Collapse
|
20
|
Mohammadi Y, Saghaei M, Saghi M, Mazhari SA, Alani B, Ehtesham N, Kenarangi T, Soosanabadi M. Significant association of functional variants in the promoter sequence of IL18 with disease susceptibility and systemic lupus erythematosus clinical parameters. Lupus 2025; 34:579-587. [PMID: 40155321 DOI: 10.1177/09612033251331256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
ObjectiveSystemic lupus erythematosus (SLE) is a chronic autoimmune disease with a complex etiology. Interleukin-18 (IL-18) possesses pro-inflammatory properties and plays a central role in the development of SLE. In this study, we assessed the association between two functional variants that affect the expression of IL-18, namely -607C > A (rs1946518) and -137G > C (rs187238), and the risk of SLE development.MethodsAs a case-control study, 251 peripheral blood samples were collected from 121 SLE patients and 130 healthy participants. Genotyping of these polymorphisms was performed using the high-resolution melting (HRM) method, which employs real-time polymerase chain reaction.ResultsOur findings revealed a significant association between the AA genotype and A allele in rs1946518, showing a decreased risk of SLE (AA vs CC; OR: 0.386; 95% CI [0.174-0.828], A vs C; OR: 0.548; 95% CI [0.369-0.809]). Analogously, the CC genotype and C allele in rs187238 exhibited a similar trend (CC vs GG; OR: 0.240; 95% CI [0.055-0.803], C vs G; OR: 0.604; 95% CI [0.390-0.928]), indicating a reduced risk of SLE Moreover, SLE subjects with the protective allele in rs1946518 (AA + AC) demonstrated significantly lower levels of CRP, and Anti-dsDNA, suggesting lower disease activity. These patients also had a later age of onset, and a lower incidence of renal involvement and creatinine levels, indicating milder disease severity (p < .05).ConclusionThe study indicates a significant relationship between the rs1946518 and rs187238 variants in IL-18 and a reduced risk of SLE. Furthermore, rs1946518 was found to be associated with certain clinical features related to disease activity and severity.
Collapse
Affiliation(s)
- Yousef Mohammadi
- Student Research Committee, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
- Fetal Health Research Center, Hope Generation Foundation, Tehran, Iran
| | - Mozhdeh Saghaei
- Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mostafa Saghi
- Student Research Committee, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
- Fetal Health Research Center, Hope Generation Foundation, Tehran, Iran
| | | | - Behrang Alani
- Autoimmune Disease Research Center, Kashan University of Medical Sciences, Kashan, Iran
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Naeim Ehtesham
- Department of Medical Genetics, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Taiebe Kenarangi
- Student Research Committee, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mohsen Soosanabadi
- Department of Medical Genetics, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
21
|
Tawara-Iida T, Usui J, Ebihara I, Ishizu T, Kobayashi M, Maeda Y, Kobayashi H, Yamagata K. Low serum free IL-18 is a novel potential marker for predicting infectious events in patients at dialysis initiation. Clin Kidney J 2025; 18:sfaf094. [PMID: 40322680 PMCID: PMC12044333 DOI: 10.1093/ckj/sfaf094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Indexed: 05/08/2025] Open
Abstract
Background Compared to the general population, individuals who are undergoing hemodialysis are at a higher risk of contracting severe infectious diseases, and their mortality rate from infectious diseases is also higher. We investigated the serum free interleukin-18 [free state of interleukin-18 (IL-18)] concentration as a prognostic factor for hemodialysis patients' infection risk. Methods The Ibaraki Dialysis Initiation Cohort (iDIC) study is a multicenter prospective cohort investigation of patients undergoing a new initiation of dialysis in a local region of Japan. We performed a survival analysis of several events requiring hospitalization and compared the Kaplan-Meier curves of the "low" and "high" serum free IL-18 concentration groups. To adjust for confounding factors, we also performed a Cox proportional hazards analysis. Results We analyzed the serum free IL-18 concentration of samples from 295 patients randomly selected from the blood sample bank of the iDIC study. The mean free IL-18 concentration was 8.7 ± 5.3 pmol/l. The cumulative incidence of infectious events was significantly higher in the low free IL-18 group (<6.0 pmol/l, log-rank test P < .01). The Cox proportional hazards analysis revealed that low serum free IL-18 (<6.0 pmol/l) was an independent factor associated with the development of infectious events. Total IL-18 and IL-18BP (binding protein) showed no association with infectious events. Conclusion A low serum free IL-18 concentration in the dialysis initiation period is a potential marker for predicting the development of severe infection in these patients.
Collapse
Affiliation(s)
- Takashi Tawara-Iida
- Department of Nephrology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Joichi Usui
- Department of Nephrology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Itaru Ebihara
- Department of Nephrology, Mito Saiseikai General Hospital, Mito, Japan
| | - Takashi Ishizu
- Department of Renal and Dialysis Medicine, Tsukuba Central Hospital, Ushiku, Japan
- Central Jin Clinic, Ryugasaki, Japan
| | - Masaki Kobayashi
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ami, Japan
| | - Yoshitaka Maeda
- Nephrology Division, Department of Internal Medicine, JA Toride Medical Center, Toride, Japan
| | - Hiroaki Kobayashi
- Department of Nephrology, Ibaraki Prefectural Central Hospital, Kasama, Japan
| | - Kunihiro Yamagata
- Department of Nephrology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
22
|
Gopalakrishnan R, Wang Y, Kapczinski F, Frey BN, Wollenhaupt-Aguiar B. Peripheral protein inflammatory biomarkers in bipolar disorder and major depressive disorder: A systematic review and meta-analysis. J Affect Disord 2025; 376:149-168. [PMID: 39894226 DOI: 10.1016/j.jad.2025.01.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 01/18/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVES Bipolar disorder (BD) and major depressive disorder (MDD) are mood disorders. The most frequent clinical presentation of BD and MDD is depression, which contributes to high rates of misdiagnosis between disorders. To support diagnostic discrimination and therapeutic stratification, we aim to perform a systematic review and meta-analysis evaluating peripheral protein inflammatory biomarkers between BD and MDD, with a focus on the depressive state. METHODS We conducted a literature search on PubMed, PsycInfo and Embase with no year/language restrictions. Original studies including human participants with a BD or MDD diagnosis which directly compared levels of peripheral protein inflammatory biomarkers between groups were included. A random effects meta-analysis was performed. RESULTS 35 studies were included in the systematic review. 9 studies were included in the meta-analysis. The meta-analysis showed IL-7 (p < 0.01) levels were significantly decreased in BD, and IL-9 (p < 0.01), CCL3 (p = 0.03), CCL4 (p = 0.01), CCL5 (p = 0.02) and CCL11 (p = 0.04) levels were significantly increased in BD. LIMITATIONS High heterogeneity and limited dataset size restricted our meta-analysis to a small subset of biomarkers and limited our exploration of the effects of moderator variables. CONCLUSION This study found differences in IL-7, IL-9, CCL3, CCL4, CCL5 and CCL11 between BD and MDD in a depressive state. These findings support the notion that inflammation is associated with mood disorder pathophysiology, particularly with respect to T-cell network dysregulation. Further studies can assist in better understanding differences between disorders and work towards clinical applications.
Collapse
Affiliation(s)
- Ridhi Gopalakrishnan
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Centre for Clinical Neurosciences, McMaster University, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Yifan Wang
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Centre for Clinical Neurosciences, McMaster University, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Flavio Kapczinski
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Centre for Clinical Neurosciences, McMaster University, St. Joseph's Healthcare, Hamilton, ON, Canada; Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Bianca Wollenhaupt-Aguiar
- Centre for Clinical Neurosciences, McMaster University, St. Joseph's Healthcare, Hamilton, ON, Canada; Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, ON, Canada.
| |
Collapse
|
23
|
Shen Y, Lin P. The Role of Cytokines in Postherpetic Neuralgia. J Integr Neurosci 2025; 24:25829. [PMID: 40302252 DOI: 10.31083/jin25829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 05/02/2025] Open
Abstract
Nerve injury is a significant cause of postherpetic neuralgia (PHN). It is marked by upregulated expression of cytokines secreted by immune cells such as tumor necrosis factor alpha, interleukin 1 beta (IL-1β), IL-6, IL-18, and IL-10. In neuropathic pain (NP) due to nerve injury, cytokines are important for the induction of neuroinflammation, activation of glial cells, and expression of cation channels. The release of chemokines due to nerve injury promotes immune cell infiltration, recruiting inflammatory cytokines and further amplifying the inflammatory response. The resulting disequilibrium in neuroimmune response and neuroinflammation leads to a reduction of nerve fibers, altered nerve excitability, and neuralgia. PHN is a typical NP and cytokines may induce PHN by promoting central and peripheral sensitization. Currently, treating PHN is challenging and research on the role of cytokine signaling pathways in PHN is lacking. This review summarizes the potential mechanisms of cytokine-mediated PHN and discusses the cytokine signaling pathways associated with the central and peripheral sensitization of PHN. By elucidating the mechanisms of cytokines, the cells and molecules that regulate cytokines, and their signaling systems in PHN, this review reveals important research developments regarding cytokines and their signaling pathways mediating PHN, highlighting new targets of action for the development of analgesic drugs.
Collapse
Affiliation(s)
- Yunyan Shen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Ping Lin
- Department of Geriatrics, Hangzhou Third People's Hospital, 310009 Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Luo Y, Li J, Fu Q, Zhang P, Song X, Liu M, Mo R, Fu J, Tang S, Wu J, Yang X, Liu X, Wang T, Ni G. Caerin 1.1 and 1.9 peptides induce acute caspase 3/GSDME-mediated pyroptosis in epithelial cancer cells. Sci Rep 2025; 15:13377. [PMID: 40251208 PMCID: PMC12008296 DOI: 10.1038/s41598-025-96438-0] [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: 08/14/2024] [Accepted: 03/28/2025] [Indexed: 04/20/2025] Open
Abstract
Caerin peptides exhibit a dual role in cancer treatment by directly killing cancer cells and modulating the tumour microenvironment to enhance anti-tumour immunity. This study investigates the mechanisms underlying caerin 1.1/1.9-induced acute cell death in epithelial cancer cells and explores their therapeutic potential. HeLa, A549, and Huh-7 cancer cell lines were treated with caerin 1.1/1.9 peptides. Morphological observations, flow cytometry, lactate dehydrogenase (LDH) release, and IL-18 secretion assays revealed the occurrence of pyroptosis following treatment. Specifically, a 1-h treatment with caerin 1.1/1.9 induced pyroptosis in HeLa, A549, and Huh-7 cells, characterised by cell swelling, membrane bubbling, and the release of IL-18 and LDH. Western blotting confirmed the upregulation of pyroptosis markers, including caspase-3, cleaved caspase-3, and GSDME-N fragments. These findings highlight the significant role of caerin peptides in inducing acute pyroptosis, a form of programmed cell death that enhances the immunogenicity of dying cancer cells, thus potentially improving the effectiveness of immunotherapies. This research underscores the therapeutic potential of caerin 1.1/1.9 peptides in cancer treatment, providing a foundation for developing new anti-cancer strategies that leverage both direct cytotoxic effects and immune modulation to achieve more effective and sustained anti-tumour responses.
Collapse
Affiliation(s)
- Yuandong Luo
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Junjie Li
- Zhongao Biomedical Technology (Guangdong) Co. Ltd, Zhongshan, 528400, Guangdong, China
| | - Quanlan Fu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Pingping Zhang
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Xinyi Song
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Mengqi Liu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Rongmi Mo
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Jiawei Fu
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Shuxian Tang
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Jialing Wu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Xiaodan Yang
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Xiaosong Liu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China.
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China.
| | - Tianfang Wang
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
| | - Guoying Ni
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China.
| |
Collapse
|
25
|
Elhani I, Calas L, Bejar F, Fautrel B, Pieroni L, Hentgen V, Mertz P, Mitrovic S, Delplanque M, Savey L, Georgin-Lavialle S. Serum interleukin-18 levels are specifically elevated in auto-inflammatory diseases involving the pyrin inflammasome: A study on 516 patients. Eur J Intern Med 2025:S0953-6205(25)00136-0. [PMID: 40204564 DOI: 10.1016/j.ejim.2025.04.002] [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: 10/11/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
Auto-inflammatory diseases (AIDs) are characterized by excessive activation of innate immunity. Current biomarkers, such as C-reactive protein (CRP) and serum Amyloid A (SAA), are not disease-specific and cannot reflect disease severity. Interleukin-18 (IL-18), a pro-inflammatory cytokine of the IL-1 superfamily, has been recently studied as, biomarker for AIDs; This study aims to evaluate total serum IL-18 levels in a large cohort of AID patients from the adult French national reference center for AID. We conducted a retrospective analysis of 708 IL-18 measurements from 516 patients. The highest IL-18 levels were observed in diseases involving the pyrin inflammasome, such as Familial Mediterranean fever, mevalonate kinase deficiency, CDC42-associated AID and PSTPIP1-associated AID. Receiver operating characteristic (ROC) curve analysis demonstrated an AUC of 0.87 for IL-18, with a sensitivity of 83.4 % and specificity of 76.2 % at a cut-off value of 412 pg/mL, in differentiating pyrin inflammasome-related diseases from other monogenic inflammatory diseases. Our findings suggest the utility of total serum IL-18 as a diagnostic tool, particularly for pyrin inflammasome-related AIDs, that could in the future help to personalized treatment strategies.
Collapse
Affiliation(s)
- Inès Elhani
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, Tenon hospital, department of internal medicine, DMU 3ID, Paris, France.
| | - Laure Calas
- Tenon Hospital, department of biochemistry, Paris, France; Sorbonne Université, INSERM UMRS 1155 UPMC, Hôpital Tenon, Paris, France
| | - Farah Bejar
- Tenon Hospital, department of biochemistry, Paris, France; Sorbonne Université, INSERM UMRS 1155 UPMC, Hôpital Tenon, Paris, France
| | - Bruno Fautrel
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, INSERM UMRS 1136, Hôpital de La Pitié Salpêtrière, Paris, France
| | | | | | - Philippe Mertz
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, Tenon hospital, department of internal medicine, DMU 3ID, Paris, France
| | - Stéphane Mitrovic
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, INSERM UMRS 1136, Hôpital de La Pitié Salpêtrière, Paris, France
| | - Marion Delplanque
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, Tenon hospital, department of internal medicine, DMU 3ID, Paris, France
| | - Léa Savey
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, Tenon hospital, department of internal medicine, DMU 3ID, Paris, France
| | - Sophie Georgin-Lavialle
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, Tenon hospital, department of internal medicine, DMU 3ID, Paris, France; INSERM U938; ERN RITA.
| |
Collapse
|
26
|
Chen PK, Wey SJ, Chen DY. Toward a personalized therapy of still's disease based on immunologic endotypes: a narrative review. Expert Rev Clin Immunol 2025; 21:401-412. [PMID: 39925174 DOI: 10.1080/1744666x.2025.2465406] [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: 10/15/2024] [Revised: 01/05/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION Accumulative evidence indicates that both innate and adaptive immunity are involved in pathogenesis of Still's disease, an autoinflammatory disease. With Increasing insights into the pathogenesis of Still's disease coupled with the availability of emerging targeted therapeutics, it may be the unmet need for personalizing therapy and achieving a treat-to-target goal. We aim to summarize the available evidence regarding immunopathogenesis of Still's disease and therapeutic strategies based on immunologic endotypes. AREAS COVERED We searched MEDLINE database using the PubMed interface and reviewed relevant English-language literature from 1971 to 2024. This review focuses on the existing evidence on pathophysiology and immunological endotypes of Still's disease and their implications for personalized strategies for patients with this disease. EXPERT OPINION Targeting the complex immunopathogenesis of Still's disease, emerging new agents are available for treatment, including biologic disease-modifying anti-rheumatic drugs (bDMARDs) and targeted synthetic DMARDs (tsDMARDs) such as Janus kinase inhibitors (JAKi). According to the updated evidence, meta-analyses, and recommendations, we propose a flow chart emphasizing personalized therapeutic strategies based on immunological endotypes. Hopefully, the therapeutic strategy might help guide the optimal selection of b/tsDMARDs to achieve a 'treat-to-target' goal in Still's disease. This proposed flow chart will be updated as newer evidence emerges.
Collapse
Affiliation(s)
- Po-Ku Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung, Taiwan
| | - Shiow-Jiuan Wey
- Division of Dermatology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
27
|
Zabeti Touchaei A, Vahidi S, Samadani AA. The Practical Landscape of Cytokine-Targeted miRNAs to Enhance NK Cell Function in Cancer Immunotherapy: A Bioinformatic Analysis. Cancer Rep (Hoboken) 2025; 8:e70192. [PMID: 40189842 PMCID: PMC11973122 DOI: 10.1002/cnr2.70192] [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: 07/09/2024] [Revised: 03/06/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
INTRODUCTION Suppression within the tumor microenvironment (TME) hampered natural killer (NK) cells and their role in cancer immunotherapy. This study explores how interleukin (IL) signaling (IL-12A, IL-12B, IL-15, IL-18) and interferon gamma (IFNG or IFN-γ) interact with microRNAs to regulate NK cell function in cancer. METHODS We identify the targeted microRNAs (miRNAs) for these genes and the key pathways influencing various cancers through comprehensive analyses, including protein-protein interaction networks, protein co-expression, miRNA targeting prediction, homology, mRNA-miRNA regulatory networks, gene set enrichment, and signaling pathway analysis. RESULTS Our analysis revealed a significant association between genes encoding interleukins and IFNG with NK cell infiltration across various cancers. Additionally, we identified several miRNAs (hsa-miR-590-3p, hsa-miR-340-5p, hsa-miR-495-3p, hsa-miR-5692a, hsa-miR-130a-3p) that potentially regulate NK cell function by targeting these genes. These miRNAs participate in critical pathways essential for NK cell function. Notably, our findings suggest a key role for mRNA-miRNA co-regulation in suppressing NK cells within the tumor microenvironment. CONCLUSION This study highlights the potential of targeting these identified miRNAs as a strategy to enhance NK cell function and improve the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
| | - Sogand Vahidi
- Medical Biology Research CenterKermanshah University of Medical SciencesKermanshahIran
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma InstituteGuilan University of Medical SciencesRashtIran
| |
Collapse
|
28
|
Li J, Luo R, Fu Y, Liu S, Dong Q, Sun Y, Tian X, Zhu Y, Wang P, Guo L, Lu Q, Ye C, Fu S, Qiu Y. Baicalin, Amoxicillin, and Probenecid Provide Protection in Mice Against Glaesserella parasuis Challenge. Biomolecules 2025; 15:507. [PMID: 40305201 PMCID: PMC12024593 DOI: 10.3390/biom15040507] [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: 02/19/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 05/02/2025] Open
Abstract
Glaesserella parasuis (G. parasuis) causes Glässer's disease and systemic inflammatory responses in the host. The currently available therapies have limited efficacy and fail to achieve a balance between anti-inflammatory and antibacterial effects. In this study, we investigated the effects of baicalin, amoxicillin, and probenecid on blood biochemical parameters, routine blood indicators, survival rate, bacterial burden, and pathological tissue damage in G. parasuis-challenged mice. Treatment with baicalin, amoxicillin, and probenecid significantly modified the blood biochemical parameters and routine blood test indicators, increased the survival rate, attenuated the bacterial burden, and alleviated pathological tissue damage in G. parasuis-challenged mice. Treatment with baicalin, amoxicillin, and probenecid also increased the number of CD3+, CD3+CD4+, and CD3+CD8+ T cells as measured by flow cytometry, and restored the intensity of the CD3, CD4, and CD8 protein expression in the blood vessels of G. parasuis-challenged mice by immunohistochemistry. These compounds reduced interleukin 1β (IL-1β), IL-18, tumor necrosis factor alpha (TNF-α), and high mobility group box 1 protein (HMGB1) expression in the spleen of G. parasuis-challenged mice. Furthermore, baicalin, amoxicillin, and probenecid inhibited activation of the family pyrin domain containing 3 (NLRP3) inflammasome and apoptosis in the spleen of G. parasuis-challenged mice. This study showed the important roles of baicalin, amoxicillin, and probenecid in the modulation of the inflammatory response of Glässer's disease. The findings might provide new strategies for combination therapy using antibiotics and anti-inflammatory drugs to control G. parasuis infection.
Collapse
Affiliation(s)
- Jingyang Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Ronghui Luo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Yunjian Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Siyu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Qiaoli Dong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Yamin Sun
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Xinyue Tian
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Yi Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Peiyi Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Ling Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Qirong Lu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| |
Collapse
|
29
|
Piekoszewska-Ziętek P, Korytowska-Przybylska N, Pańczyk-Tomaszewska M, Olczak-Kowalczyk D. Salivary Interleukin-6 and Interleukin-18 Levels and Their Association with Dental Health in Children with Idiopathic Nephrotic Syndrome. Int J Mol Sci 2025; 26:3175. [PMID: 40243946 PMCID: PMC11989940 DOI: 10.3390/ijms26073175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Idiopathic nephrotic syndrome (NS) is associated with immune dysfunction and increased susceptibility to infections. Oral health may influence systemic inflammation and disease progression. This study aimed to evaluate the salivary levels of interleukin-6 (IL-6) and interleukin-18 (IL-18) in children with NS and their association with dental health, particularly caries prevalence and the consequences of untreated caries. A cross-sectional study was conducted on 86 children aged 5-17 years, including 40 NS patients and 46 healthy controls. Clinical dental examinations assessed caries prevalence using the dmft/DMFT index and the impact of untreated caries using the pufa/PUFA index. Unstimulated saliva samples were collected, and IL-6 and IL-18 concentrations were measured using enzyme-linked immunosorbent assay. NS patients exhibited a significantly lower prevalence of active carious lesions than controls (50% vs. 72%, p = 0.039). The DMFT index was lower in the NS group (p = 0.003). Salivary IL-6 levels were significantly reduced in NS patients compared to controls (p = 0.015), while IL-18 levels showed no significant difference. IL-6 positively correlated with decayed permanent teeth and pulp/periapical tissue diseases, whereas IL-18 correlated with white spot lesions and pulp infections. IL-6 and IL-18 could serve as potential non-invasive indicators of disease progression in NS patients.
Collapse
Affiliation(s)
| | - Natalia Korytowska-Przybylska
- Department of Drug Chemistry, Pharmaceutical and Biomedical Analysis, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | | | | |
Collapse
|
30
|
Leng L, Li Y, Xu T, Shen J, Li L, Li X. Causal Association Between Circulating Inflammatory Proteins and Autoimmune Liver Disease: a Bidirectional Two-Sample Mendelian Randomization Study. Immunotargets Ther 2025; 14:279-289. [PMID: 40161480 PMCID: PMC11955185 DOI: 10.2147/itt.s508140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction To investigate whether there is a direct causal relationship between circulating inflammatory proteins and autoimmune liver disease (AILD). Materials and Methods We collected genetic data for various AILD from the Genome Wide Association Studies (GWAS) dataset. The latest research provides GWAS data for 91 proteins associated with inflammation. Perform bidirectional two sample Mendelian randomization (MR) analysis using inverse variance weighted (IVW) to determine the causal relationship between inflammatory proteins and AILD, and use Mendelian randomization Egger method (MR Egger), weighted median (WM), and weighted mode as supplementary evaluations. In addition, we conducted sensitivity analysis. Results Positive MR analysis showed that CDCP1 (OR=1.363, p=0.0465) and IL-18 (OR=1.416, p=0.0477) were associated with higher including autoimmune hepatitis (AIH) risk. Higher CXCL11 (OR=1.574, p=9.23×10-5) were associated with an increased risk of primary biliary cholangitis (PBC). Lower levels of three inflammatory proteins were associated with increased risk of PBC. TNFSF12 (OR=1.827, p=0.0001, p_adj_fdr=0.0063), CD6 isoform (OR=1.126, p=0.0389), CCL20 (OR=1.880, p=0.0395) are associated with increased risk of primary sclerosing cholangitis (PSC). Reverse MR imaging showed that PBC may promote the expression levels of CCL4 (OR=1.023, p=0.0201) and OSM (OR=1.022, p=0.0236). PSC may promote the expression of five inflammatory proteins. Sensitivity analysis further excluded the effects of heterogeneity and horizontal pleiotropy. Conclusion This study indicates a potential association between circulating inflammatory proteins and AILD, which may become a new diagnostic indicator or drug target for clinical application in the prevention and treatment of AILD. However, further investigation is needed.
Collapse
Affiliation(s)
- Lina Leng
- Department of Rheumatology, Xingtai People’s Hospital, Xingtai, 054001, Hebei Province, People’s Republic of China
| | - Ying Li
- Department of Oncology, 82 Group Hospital of Chinese People’s Liberation Army, Baoding, Hebei Province, 071000, People’s Republic of China
| | - Tao Xu
- Department of Internal Medicine, Graduate School of Hebei North University, Zhangjiakou, Hebei Province, 075000, People’s Republic of China
| | - Jingfang Shen
- Department of Rheumatology, Xingtai People’s Hospital, Xingtai, 054001, Hebei Province, People’s Republic of China
| | - Lianju Li
- Department of Rheumatology, Xingtai People’s Hospital, Xingtai, 054001, Hebei Province, People’s Republic of China
| | - Xiaoli Li
- Department of Rheumatology, Xingtai People’s Hospital, Xingtai, 054001, Hebei Province, People’s Republic of China
| |
Collapse
|
31
|
Alboni S, Tascedda F, Uezato A, Sugama S, Chen Z, Marcondes MCG, Conti B. Interleukin 18 and the brain: neuronal functions, neuronal survival and psycho-neuro-immunology during stress. Mol Psychiatry 2025:10.1038/s41380-025-02951-z. [PMID: 40121365 DOI: 10.1038/s41380-025-02951-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/27/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Interleukin 18 (IL-18) is a pleiotropic cytokine that regulates peripheral innate and adaptive immune response and is also expressed in the brain. Here, we summarize the current knowledge on the biology of IL-18 in the brain and the efforts to determine its significance concerning neurological and psychiatric conditions. The picture that emerges is that of a heavily regulated molecule that can contribute to neuroinflammatory-mediated neuronal survival but can also serve as a neuromodulator that affects behaviour. We also summarize evidence showing how the brain can control the synthesis of peripheral IL-18 during stress by hormonal and neuronal signalling, regulating tissue-specific promoter usage. We discuss how this may represent one of the mechanisms by which the brain affects immune functions and what its implications are when considering IL-18 as a biomarker of psychiatric conditions.
Collapse
Affiliation(s)
- Silvia Alboni
- University of Modena and Reggio Emilia, Department of Life Sciences via Campi 287, 41125, Modena, Italy
| | - Fabio Tascedda
- University of Modena and Reggio Emilia, Department of Life Sciences via Campi 287, 41125, Modena, Italy
| | - Akihito Uezato
- Center for Basic Medical Research, International University of Health and Welfare, 2600-1 Kitakanemaru, Otawara, Tochigi, 324-8501, Japan
| | - Shuei Sugama
- Center for Basic Medical Research, International University of Health and Welfare, 2600-1 Kitakanemaru, Otawara, Tochigi, 324-8501, Japan
| | - Zuxin Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, P. R. China
| | | | - Bruno Conti
- San Diego Biomedical Research Institute, 3525 John Hopkins Ct, San Diego, CA, 92121, USA.
| |
Collapse
|
32
|
Okoye GD, Kumar A, Ghanbari F, Chowdhury NU, Wu L, Newcomb DC, Van Kaer L, Algood HMS, Joyce S. Single-cell map of innate-like lymphocyte response to Francisella tularensis infection reveals interleukin-17-dependent protection by MAIT cells. iScience 2025; 28:111810. [PMID: 40160424 PMCID: PMC11951026 DOI: 10.1016/j.isci.2025.111810] [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: 05/31/2024] [Revised: 08/07/2024] [Accepted: 01/10/2025] [Indexed: 04/02/2025] Open
Abstract
Early immune dynamics during the initiation of fatal tularemia caused by Francisella tularensis infection remain unknown. Unto that end, we generated a transcriptomic map at single-cell resolution of the innate-like lymphocyte responses to F. tularensis live vaccine strain (LVS) infection of mice. We found that both interferon-γ (IFN-γ)-producing type 1 and interleukin-17 (IL-17)-producing type 3 innate-like lymphocytes expanded in the infected lungs. Natural killer (NK) and NKT cells drove the type 1 response, whereas mucosal-associated invariant T (MAIT) and γδ T cells drove the type 3 response. Furthermore, tularemia-like disease resistant NKT cell-deficient, Cd1d -/- mice accumulated more MAIT1 cells, MAIT17 cells, and cells with a hybrid phenotype between MAIT1 and MAIT17 cells than wild-type mice. Critically, adoptive transfer of LVS-activated MAIT cells from Cd1d -/- mice, which were enriched in MAIT17 cells, was sufficient to protect LVS-susceptible, immunodeficient RAG2 -/- mice from severe LVS infection-inflicted pathology. Collectively, our findings position MAIT cells as potential mediators of IL-17-dependent protection from pulmonary tularemia-like disease.
Collapse
Affiliation(s)
- G. Donald Okoye
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Nashville, TN 37232, USA
| | - Amrendra Kumar
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology & Inflammation, Nashville, TN 37232, USA
| | - Farshad Ghanbari
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Nowrin U. Chowdhury
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Nashville, TN 37232, USA
| | - Lan Wu
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology & Inflammation, Nashville, TN 37232, USA
| | - Dawn C. Newcomb
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology & Inflammation, Nashville, TN 37232, USA
| | - Luc Van Kaer
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology & Inflammation, Nashville, TN 37232, USA
| | - Holly M. Scott Algood
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology & Inflammation, Nashville, TN 37232, USA
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sebastian Joyce
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology & Inflammation, Nashville, TN 37232, USA
| |
Collapse
|
33
|
Deng Y, Wang Y, Cheng Y, Lei M, Luo Y, Gu W, Cai B, Li L, Zhang N. Expression of interleukin-18 in primary Sjögren syndrome and its potential mechanisms with disease: A systematic review and meta-analysis. Medicine (Baltimore) 2025; 104:e41919. [PMID: 40128067 PMCID: PMC11936574 DOI: 10.1097/md.0000000000041919] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/02/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Interleukin-18 (IL-18), an IL-1 family cytokine with potent ability to induce interferon gamma production and enhance Th1 response, was elevated in a group of autoimmune diseases including primary Sjögren syndrome (pSS). IL-18 dysregulation can contribute to the pathogenesis of these disorders by exerting function in innate and adaptive immunity. This study is aimed at comparing the level of IL-18 in pSS patients and explored the association between IL-18 and pSS. METHODS Six databases, including PubMed, Web of Science, Embase, Ovid Medline, Scopus and China National Knowledge Infrastructure, were searched. The quality of the included studies was assessed using the Newcastle-Ottawa Scale criteria. We analyzed IL-18 concentrations in the serum and tear of pSS patients and healthy controls (HCs), and review the potential mechanism between IL-18 and pSS. RESULTS Eleven articles were included in this study, which reported IL-18 levels in serum and tear of pSS patients and HCs. IL-18 levels in pSS patients were significantly higher than those in HCs (standard mean difference = 1.28, 95% confidence interval 0.75-1.82, P < .001), with substantial heterogeneity (I2 = 90%, P < .001) among the studies. The level of IL-18 in serum was analyzed separately and was also higher than those in HCs (standard mean difference = 1.28, 95% confidence interval 0.68-1.88, P < .001), with significant heterogeneity (I2 = 91%, P < .001). Apart from that, the potential pathogenesis of pSS was concluded comprehensively. CONCLUSION IL-18 abundance was evidently elevated in pSS patients and can thus likely be used as a reliable biomarker to monitor and track the progression of pSS, and further to become a good target for treatment of pSS patient.
Collapse
Affiliation(s)
- Ying Deng
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yijue Wang
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yijuan Cheng
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- West China Hospital, Sichuan University, Chengdu, China
| | - Min Lei
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yingyu Luo
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wenling Gu
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Boyu Cai
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Division of Renal and Endocrinology, Qin Huang Hospital, Xi’an, China
| | - Nannan Zhang
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Chen X, Zhang R, Xie H, Li S, Guo J, Wang Y. Association of the IL-10 and IL-18 polymorphisms with nasopharyngeal carcinoma risk. Front Oncol 2025; 15:1543182. [PMID: 40115022 PMCID: PMC11922698 DOI: 10.3389/fonc.2025.1543182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
Objective To evaluate the possible association of the cytokine polymorphisms with the risk of nasopharyngeal carcinoma (NPC). Methods We performed a comprehensive search of electronic databases from PubMed, Web of Science, Embase, and CNKI. Articles related to the cytokine polymorphisms in patients with NPC and healthy controls from inception to 1 April 2024 were included. The results were analysed independently by two reviewers using RevMan 5.4 software. Summary odds ratio (OR) and 95% confidence interval (CI) were used to evaluate cancer risk. Results Our results showed that IL-10 1082A>G showed a significant difference only in the Dominant model, but in the Asian population, a significant difference was shown in all models. IL-18 607C>A polymorphism showed significant differences in the Allele model, Heterozygote model, and Homozygote model. In addition, the IL-18 137G>C polymorphism showed significant differences in all models. No statistically significant association was found between IL-8 251A>T, IL-10 819T>C polymorphism, and the risk of NPC. Conclusion Our meta-analysis results suggest that the IL-18 607C>A and IL-18 137G>C polymorphism are associated with the increased risk of NPC, and IL-10-1082 A/G polymorphism is associated with the increased risk of NPC in Asian populations.
Collapse
Affiliation(s)
- Xueru Chen
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Ruibin Zhang
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
| | - Hui Xie
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
| | - Sha Li
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Jincai Guo
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yan Wang
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
35
|
Arul Arasan TS, Jorgensen R, Van Antwerp C, Ng PKW, Gangur V. Advances in Mechanisms of Anaphylaxis in Wheat Allergy: Utility of Rodent Models. Foods 2025; 14:883. [PMID: 40077585 PMCID: PMC11899146 DOI: 10.3390/foods14050883] [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: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Wheat is a staple and nutritious food that is consumed globally. However, it is identified as a major allergenic food because of its capacity to trigger life-threatening systemic anaphylaxis. The specific mechanisms that underlie this systemic anaphylaxis in wheat allergy are incompletely understood. As a result, several rodent models have been developed to study anaphylaxis in wheat allergies. In this paper, we have conducted a comprehensive review of wheat-induced anaphylaxis using Google Scholar and PubMed databases with relevant keywords. The following objectives were addressed: (1) to determine the complexity of wheat-induced anaphylaxis; (2) to summarize the role of genetic susceptibility in wheat anaphylaxis; (3) to identify the environmental factors involved in the development of wheat anaphylaxis; (4) to map the current status of mechanisms involved in wheat anaphylaxis; (5) to identify the approaches, strengths, and limitations of rodent models of wheat anaphylaxis; and (6) to identify challenges and opportunities in this area of science. Our findings provide a comprehensive updated critical resource for the future research agenda in wheat allergy-associated anaphylaxis, particularly using rodent models as attractive pre-clinical tools.
Collapse
Affiliation(s)
- Tamil Selvan Arul Arasan
- Food Allergy and Immunology Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; (T.S.A.A.); (R.J.); (C.V.A.)
| | - Rick Jorgensen
- Food Allergy and Immunology Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; (T.S.A.A.); (R.J.); (C.V.A.)
| | - Chris Van Antwerp
- Food Allergy and Immunology Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; (T.S.A.A.); (R.J.); (C.V.A.)
| | - Perry K. W. Ng
- Cereal Science Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48823, USA;
| | - Venu Gangur
- Food Allergy and Immunology Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; (T.S.A.A.); (R.J.); (C.V.A.)
| |
Collapse
|
36
|
Sousa CH, Mercier M, Rioux‐Leclercq N, Flecher E, Bendavid C, Val‐Laillet D, Ferrant J, Jaillard S, Loiseau E, Branchereau J, Berkane Y, Nyangoh Timoh K, Carton I, Le Lous M, Lavoue V, Dion L. Hypothermic machine perfusion in uterus transplantation in a porcine model: A proof of concept and the first results in graft preservation. Acta Obstet Gynecol Scand 2025; 104:461-473. [PMID: 39868864 PMCID: PMC11871101 DOI: 10.1111/aogs.15056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Graft optimization is a necessity in order to develop uterus transplantation from brain-dead donors, as a complement to living donors, as these grafts are rare and the last organs retrieved in multiple organ donation. The aim of this study was to assess the feasibility and interest of hypothermic machine perfusion (HMP) in uterus transplantation using a porcine model; secondary outcomes were the evaluation of the graft's tolerance to a prolonged cold ischaemia time and to find new biomarkers of uterus viability. MATERIAL AND METHODS Fifteen uterus allotransplantations were performed in a porcine model, after 18 h of cold ischaemia, divided in three groups: Static cold storage in a HTK solution, HMP (with the VitaSmart (™) machine Bridge to Life Ltd.) with a UW-MP solution, and static cold storage in a UW solution. The main outcome was macroscopic: uterine arteries pulsatility, recoloration, and bleeding at the cut. Secondary outcomes were histological analyses (Zitkute and inflammation scores), caspase3 immunohistochemistry and plasmatic dosage of biomarkers. RESULTS 14/15 allotransplantations were performed according to the protocol and met the criteria of macroscopic vitality. Grafts treated with HMP (MP did not show significantly more tissue) damage than the recipient's uterus, contrary to grafts in static cold storage, independently of the solution used. This difference disappeared one and 3 h after uterus transplantation. Plasma dosages before and after uterus transplantation did not allow to identify a new biomarker of uterus viability. CONCLUSIONS HMP is feasible in a porcine model, without inflicting damage on the grafts during cold ischaemia time. Grafts exposed to HMP seemed to better endure reperfusion phenomena, but this advantage did not last over time.
Collapse
Affiliation(s)
- Carla Héléna Sousa
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
| | - Marion Mercier
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
| | - Nathalie Rioux‐Leclercq
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
- Department of PathologyRennes University Hospital, Hôpital PontchaillouFrance
| | - Erwan Flecher
- Department of Cardiothoracic and Vascular SurgeryRennes University Hospital, Hôpital PontchaillouRennesFrance
| | - Claude Bendavid
- Laboratory of BiochemistryRennes University Hospital, Hôpital PontchaillouRennesFrance
| | - David Val‐Laillet
- INRAE, INSERMUniversity Of Rennes, NuMeCan Institute, Nutrition Metabolisms And CancerRennesFrance
| | - Juliette Ferrant
- Unité Mixte de Recherche (UMR)1236, Université RennesINSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
| | - Sylvie Jaillard
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
- Department of Cytogenetics and Cellular BiologyRennes University HospitalRennesFrance
| | - Emma Loiseau
- Department of GynecologyCHU Nantes Hôtel DieuNantesFrance
| | | | - Yanis Berkane
- Unité Mixte de Recherche (UMR)1236, Université RennesINSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
- Department of Plastic, Reconstructive and Aesthetic SurgeryRennes University Hospital, Hôpital SudRennesFrance
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Isis Carton
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
| | - Maëla Le Lous
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
| | - Vincent Lavoue
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
| | - Ludivine Dion
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
| |
Collapse
|
37
|
Khan J, Sadie-Van Gijsen H, Kotzé-Hörstmann LM, Kotze SH, Layman-Lemphane JI. Characterisation of the influence of dietary fat and sugar on bone health utilising densitometry, micro-computed tomography and histomorphometry. Bone 2025; 192:117380. [PMID: 39710129 DOI: 10.1016/j.bone.2024.117380] [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: 07/18/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Obesogenic feeding can affect systemic metabolism and impact bone health and microarchitecture, but the findings of published studies often appear contradictory. This study aimed to compare the effects of a medium-fat/high-sugar (MF/HS) and a high-fat/high-fructose (HF/Fr) diet on the femora of weanling male Wistar rats, examining bone mineral content and density (BMC, BMD), cortical and cancellous bone microarchitecture and the cell populations within bone. Furthermore, we explored the correlations between circulating bone-targeting factors (in particular leptin, adiponectin and insulin) and bone parameters. Rats were assigned to one of three dietary groups (control: CON; MF/HS: OB1; HF/Fr: OB2; n = 12 each) for 17 weeks. Right-hand side femora were subjected to densitometry to measure BMC and BMD, and micro-computed tomography (μCT) was utilised to assess cortical and cancellous bone. Osteoblast (N.Ob), osteoclast (N.Oc), adipocyte (N.Ad) and chondrocyte numbers (N.Ch) were quantified histomorphometrically. Diet OB1 was largely beneficial to bone, while diet OB2 exerted detrimental effects on BMC, BMD, bone microarchitecture and bone cell populations. In cortical bone, N.Ob was positively correlated with BMD, cortical area and serum leptin. In cancellous bone, N.Ob was positively correlated with serum leptin and BMD, while N.Oc was negatively correlated with serum leptin. Overall, these findings support a role for endogenous circulating leptin in promoting bone formation. We conclude that the impact of different obesogenic diets may be driven by individual dietary effects on circulating factors, which may partly explain the contradictory reports in existing literature on the impact of HF and HS diets on bone.
Collapse
Affiliation(s)
- J Khan
- Division of Clinical Anatomy, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa.
| | - H Sadie-Van Gijsen
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa.
| | - L M Kotzé-Hörstmann
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa; Division of Sport and Exercise Medicine (DiSEM), Department of Exercise, Sport and Lifestyle Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa.
| | - S H Kotze
- Division of Clinical Anatomy, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa; Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis.
| | - J I Layman-Lemphane
- Division of Clinical Anatomy, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa.
| |
Collapse
|
38
|
Nashtahosseini Z, Eslami M, Paraandavaji E, Haraj A, Dowlat BF, Hosseinzadeh E, Oksenych V, Naderian R. Cytokine Signaling in Diabetic Neuropathy: A Key Player in Peripheral Nerve Damage. Biomedicines 2025; 13:589. [PMID: 40149566 PMCID: PMC11940495 DOI: 10.3390/biomedicines13030589] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a debilitating complication of diabetes mellitus, characterized by progressive nerve damage driven by chronic hyperglycemia and systemic inflammation. The pathophysiology of DPN is significantly influenced by pro-inflammatory cytokines, such as IL-1β, IL-6, and TNF-α. These cytokines promote oxidative stress, vascular dysfunction, and neuronal degeneration by activating important signaling pathways including NF-κB and MAPK. While IL-6 promotes a pro-inflammatory microenvironment, increasing neuronal damage and neuropathic pain, TNF-α and IL-1β worsen Schwann cell failure by compromising axonal support and causing demyelination. Immune cell infiltration and TLR activation increase the inflammatory cascade in DPN, resulting in a persistent neuroinflammatory state that sustains peripheral nerve injury. The main characteristics of DPN are axonal degeneration, decreased neurotrophic support, and Schwann cell dysfunction, which weaken nerve transmission and increase susceptibility to damage. Advanced glycation end-products, TNF-α, and CXCL10 are examples of biomarkers that may be used for early diagnosis and disease progression monitoring. Additionally, crucial molecular targets have been found using proteomic and transcriptome techniques, enabling precision medicine for the treatment of DPN. This review emphasizes the importance of cytokine signaling in the pathogenesis of DPN and how cytokine-targeted treatments might reduce inflammation, restore nerve function, and improve clinical outcomes for diabetic patients.
Collapse
Affiliation(s)
| | - Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran;
| | - Elham Paraandavaji
- Clinical Research Development Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran 13399-73111, Iran
| | - Alireza Haraj
- Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran 14496-1453, Iran
| | - Bahram Fadaee Dowlat
- Faculty of Medicine, Iran University of Medical Sciences, Tehran 14496-1453, Iran
| | - Ehsan Hosseinzadeh
- Department of Surgery, School of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | | | - Ramtin Naderian
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
39
|
Lin Y, Zhang X, Sun D, Wang Q, Dou S, Zhou Q. Decoding the corneal immune microenvironment in healthy and diabetic mice during corneal wound healing. Ocul Surf 2025; 37:68-79. [PMID: 40023495 DOI: 10.1016/j.jtos.2025.02.010] [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: 10/09/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Diabetic keratopathy (DK) is an underdiagnosed ocular complication of diabetes mellitus. The changes of ocular immune microenvironment contribute to the pathogenesis of DK, while precise mechanisms remain inadequately understood. Here, we employed single-cell RNA sequencing (scRNA-seq) to elucidate the transcriptional alterations of immune cells from diabetic and healthy control mouse corneas during homeostasis and wound healing. Unbiased clustering analysis unveiled 3 major cell subsets and 11 subdivided cell clusters, including T cells, monocyte lineages, and neutrophil subpopulations. The further sub-clustering analysis demonstrated that T cells exhibited cytotoxicity characteristics in both homeostasis and wound healing of diabetic cornea. Moreover, dendritic cells preferred the migratory and maturation phenotype and may recruit and maintain cytotoxic T cells. Macrophages in diabetic cornea preferred the pro-inflammatory M1 phenotype. Under injury conditions, diabetic corneal neutrophils exhibited a more mature and functional possession of neutrophil extracellular traps (NETs). Furthermore, cell-cell communication revealed that the immune cells exhibited hyperactivation and pro-inflammatory responses, while the monocyte lineages exhibited the activating effect on T cells in diabetic cornea. This study represents the inaugural effort to establish a comprehensive scRNA-Seq transcriptomic profile of corneal immune cells during wound healing in healthy and diabetic mice, which offers a valuable reference for subsequent investigations into the pathological roles of immune cells in DK.
Collapse
Affiliation(s)
- Yujing Lin
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Xiaowen Zhang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Di Sun
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Qun Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.
| |
Collapse
|
40
|
Tian Y, Budka J, Locke FL, Westin JR, To C, Tiwari G, Mao D, Bedognetti D, Shen RR, Andrade J, Filosto S. Tumor gene expression signatures associated with outcome in large B-cell lymphoma treated with CD19-directed CAR T-cell therapy (axicabtagene ciloleucel). Front Oncol 2025; 15:1519473. [PMID: 40083872 PMCID: PMC11903469 DOI: 10.3389/fonc.2025.1519473] [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: 10/29/2024] [Accepted: 02/03/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction CAR T cell therapy provided transformative outcomes for patients with B-cell lymphoma; however, a large fraction of patients remains at risk for relapse, underlying the need to uncover mechanisms of resistance and predictive biomarkers. Herein, we leveraged the ZUMA-7 phase III randomized trial of relapsed/refractory large B-cell lymphoma (LBCL) patients treated with axicabtagene ciloleucel (axi-cel; CD19-targeting CAR T cells) to discover tumor gene expression signatures (GES) associated with outcome. Methods With tumor transcriptomics from 134 axi-cel patients, we employed multivariate penalized Cox models analyzing event-free survival (EFS), progression-free survival (PFS), and duration of response (DOR). Results and Discussion We identified two novel GES, a six-gene/transcript signature (6-GES; CD19, CD45RA, CCL22, KLRK1, SOX11, SIGLEC5) correlated with improved outcome after axi-cel (HR: 0.27, 95% CI: 0.16-0.44 for EFS), representing lymphomas with abundant target antigen (CD19) expression, adhesion molecules, and relatively low immune infiltration mostly composed of cytotoxic lymphocytes (T and NK cells) and DCs, and secondly, a 17-gene/transcript signature (17-GES; CD45RO, BCL2, IL-18R1, TNFSF4 [OX40L], KLRB1 [CD161], KIR3DL2, ITGB8, DUSP5, GPC4, PSMB5, RPS6KB1, SERPINA9, NBN,GLUD1, ESR1, ARID1A, and SLC16A1) correlated with disease progression after axi-cel (HR: 6.12, 95% CI: 3.57-10.50 for EFS), consistent with high immune inflammation and escape mechanisms, such as the upregulation of genes involved in repair of damaged DNA or chromatin remodeling, inhibition of apoptosis, and a metabolically restrictive environment. These signatures did not correlate with outcome in the standard-of-care arm of ZUMA-7 (chemotherapy, followed by transplant) or frontline therapy, supporting their predictive rather than prognostic value. The findings were technically reproduced in a subset of ZUMA-7 samples profiled by RNA-seq (axi-cel, n=124; SOC, n=125). The 6-GES was reduced, whereas the 17-GES was elevated at progression post axi-cel, consistent with the notion that these signatures represent features relevant for response and resistance to CAR T-cell therapy. Conclusion Our transcriptomic analysis identified gene expression signatures potentially predictive of outcome with CD19-directed CAR T-cell therapy, and these findings are informative for risk stratification and development of next-generation products.
Collapse
Affiliation(s)
- Yuan Tian
- Kite, a Gilead Company, Santa Monica, CA, United States
| | - Justin Budka
- Kite, a Gilead Company, Santa Monica, CA, United States
| | - Frederick L. Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, United States
| | - Jason R. Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christina To
- Kite, a Gilead Company, Santa Monica, CA, United States
| | | | - Daqin Mao
- Kite, a Gilead Company, Santa Monica, CA, United States
| | | | - Rhine R. Shen
- Kite, a Gilead Company, Santa Monica, CA, United States
| | - Jorge Andrade
- Kite, a Gilead Company, Santa Monica, CA, United States
| | | |
Collapse
|
41
|
Cocksedge SP, Mantecón L, Castaño E, Infante C, Bailey SJ. The Potential of Superoxide Dismutase-Rich Tetraselmis chuii as a Promoter of Cellular Health. Int J Mol Sci 2025; 26:1693. [PMID: 40004157 PMCID: PMC11855123 DOI: 10.3390/ijms26041693] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Tetraselmis chuii (T. chuii) is a green, marine, eukaryotic, microalgae that was authorized in the European Union (EU) as a novel food for human consumption in 2014, and as a food supplement in 2017. This narrative review will provide an overview of preclinical and clinical trials assessing the efficacy of a T. chuii-derived ingredient, characterized by a high superoxide dismutase (SOD) activity (SOD-rich T. chuii), to improve various aspects of cellular health. Collectively, results from in vitro, and more importantly in vivo research, support SOD-rich T. chuii as a potential promoter of cellular health. Principally, the ingredient appears to function as an indirect antioxidant by boosting intracellular antioxidant systems. Moreover, it can positively modulate inflammatory status by up-regulating anti-inflammatory and down-regulating pro-inflammatory cytokines and factors. In addition, SOD-rich T. chuii appears to promote cellular health though protecting from DNA damage, boosting immune function, strengthening cell structure and integrity, and positively modulating cell signaling pathways. There is also some evidence to suggest that SOD-rich T. chuii may improve aspects of mitochondrial function through the up-regulation of genes linked to mitochondrial biogenesis and ATP synthesis. From the trials conducted to date, transcriptional activation of nuclear factor erythroid 2-related factor 2 (NRF2) and sirtuin 1 (SIRT1) appear to be important in mediating the effects of SOD-rich T. chuii on cellular health. These exciting preliminary observations suggest that SOD-rich T. chuii may represent a natural blue food supplement with the potential to enhance various aspects of cellular health.
Collapse
Affiliation(s)
- Stuart P. Cocksedge
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
- Centre for Physical Activity, Sport and Exercise Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Lalia Mantecón
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Enrique Castaño
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Carlos Infante
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Stephen J. Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
| |
Collapse
|
42
|
D’Amico RC, Nagashima S, Carstens LB, Bertoldi KDG, Mataruco S, Honório D’Agostini JC, Hlatchuk EC, da Silva SB, de Noronha L, Baena CP. COVID-19 Induces Greater NLRP3 Inflammasome Activation in Obese Patients than Other Chronic Illnesses: A Case-Control Study. Int J Mol Sci 2025; 26:1541. [PMID: 40004007 PMCID: PMC11855377 DOI: 10.3390/ijms26041541] [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: 12/23/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity has been identified as an independent risk factor for severe COVID-19 unfavorable outcomes. Several factors, such as increased ACE2 receptor expression and chronic inflammation, can contribute to this relationship, yet the activation of the NLRP3 inflammasome pathway is also a key element. Our primary goal was to determine whether chronic NLRP3 inflammasome activation in people with obesity is different in critical COVID-19 and in critical chronic conditions. A retrospective analysis was conducted using clinical data and post-mortem lung tissue samples from 14 COVID-19 patients with obesity (group A) and 9 patients with obesity who died from non-COVID-19 causes (group B). Immunohistochemical analysis assessed twelve markers related to the NLRP3 inflammasome pathway. Group A showed a significantly higher expression of ASC (p = 0.0387) and CASP-1 (p = 0.0142). No significant differences were found for IL-8, TNF-α, NF-kB, NLRP3, IL-1β, and gasdermin-D. Group B had higher levels of IL-6 (p < 0.0001), IL-18 (p = 0.002), CASP-9 (p < 0.0001), and HIF (p = 0.0327). We concluded that COVID-19 activates the NLRP3 inflammasome pathway, possibly leading to pyroptotic cell death mediated by caspase-1. In contrast, people with obesity without COVID-19, despite exhibiting some markers of the NLRP3 inflammasome, are more likely to experience necroptosis mediated by caspase-9.
Collapse
Affiliation(s)
- Raíssa Campos D’Amico
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Seigo Nagashima
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Lucas Baena Carstens
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Karina de Guadalupe Bertoldi
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Sabrina Mataruco
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | | | - Elisa Carolina Hlatchuk
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Sofia Brunoro da Silva
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Lucia de Noronha
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Cristina Pellegrino Baena
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| |
Collapse
|
43
|
Wu Q, Zeng Y, Geng K, Guo M, Teng FY, Yan PJ, Lei Y, Long Y, Jiang ZZ, Law BYK, Xu Y. The role of IL-1 family cytokines in diabetic cardiomyopathy. Metabolism 2025; 163:156083. [PMID: 39603339 DOI: 10.1016/j.metabol.2024.156083] [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: 08/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Diabetic cardiomyopathy (DCM) is the primary cause of heart failure in patients with diabetes and is characterised by contractile dysfunction and left ventricular hypertrophy. The complex pathological and physiological mechanisms underlying DCM have contributed to a limited number of available treatment options. A substantial body of evidence has established that DCM is a low-grade inflammatory cardiovascular disorder, with the interleukin-1 (IL-1) family of cytokines playing crucial roles in initiating inflammatory responses and shaping innate and adaptive immunity. In this review, we aim to provide an overview of the underlying mechanisms of the IL-1 family and their relevance in DCM of various aetiologies. Furthermore, we highlighted potential therapeutic targets within the IL-1 family for the management of DCM.
Collapse
Affiliation(s)
- Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Pathology, and Luzhou Key Laboratory of Precision Pathology Diagnosis for Serious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Kang Geng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Plastic and burns surgery, National Key Clinical Construction Specialty, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Man Guo
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fang-Yuan Teng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pi-Jun Yan
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yi Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yang Long
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China.
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
44
|
Marsh RA, Bleesing JJ, Chiang SCC. Diagnostic testing for hemophagocytic lymphohistiocytosis. J Immunol Methods 2025; 537:113816. [PMID: 39855542 DOI: 10.1016/j.jim.2025.113816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 11/17/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a rare clinical syndrome caused by severe systemic hyperinflammation. HLH can be rapidly fatal if unrecognized or inadequately treated. It is important that clinicians are able to utilize diagnostic testing to assess for HLH and determine the underlying causes including possible inborn errors of immunity (IEI). This article summarizes many of the tools available to aid with the diagnostic evaluation of patients with possible HLH and underlying IEI.
Collapse
Affiliation(s)
- Rebecca A Marsh
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States; Pharming Healthcare, Warren, NJ, United States.
| | - Jack J Bleesing
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Samuel Cern Cher Chiang
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
45
|
Ormerod MBEG, Ueland T, Aas M, Hjell G, Rødevand L, Sæther LS, Lunding SH, Johansen IT, Mlakar V, Andreou D, Ueland T, Lagerberg TV, Melle I, Djurovic S, Andreassen OA, Steen NE. Limited evidence of association between dysregulated immune marker levels and telomere length in severe mental disorders. Acta Neuropsychiatr 2025; 37:e4. [PMID: 39844366 DOI: 10.1017/neu.2024.62] [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] [Indexed: 01/24/2025]
Abstract
OBJECTIVE Accelerated ageing indexed by telomere attrition is suggested in schizophrenia spectrum- (SCZ) and bipolar disorders (BD). While inflammation may promote telomere shortening, few studies have investigated the association between telomere length (TL) and markers of immune activation and inflammation in severe mental disorders. METHODS Leucocyte TL defined as telomere template/amount of single-copy gene template (T/S ratio), was determined in participants with SCZ (N = 301) or BD (N = 211) and a healthy control group (HC, N = 378). TL was analysed with linear regressions for associations with levels of 12 immune markers linked to SCZ or BD. Adjustments were made for a broad range of potential confounding variables. TL was measured by quantitative polymerase chain reaction (qPCR) and the immune markers were measured by enzyme immunoassays. RESULTS A positive association between levels of soluble tumour necrosis factor receptor 1A (sTNF-R1) and TL in SCZ (β = 0.191, p = 0.012) was observed. Plasma levels of the other immune markers were not significantly associated with TL in the BD, SCZ or HC groups. CONCLUSION There was limited evidence of association between immune markers and TL in SCZ and BD. The results provide little support for involvement of immune dysregulation, as reflected by current systemic markers, in telomere attrition-related accelerated ageing in severe mental disorders.
Collapse
Affiliation(s)
- Monica B E G Ormerod
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Monica Aas
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, England, UK
- Department of Behavioural Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Gabriela Hjell
- Department of Psychiatry, Ostfold Hospital, Graalum, Norway
| | - Linn Rødevand
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Linn Sofie Sæther
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | | | | | - Vid Mlakar
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Dimitrios Andreou
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
- Division of Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| | - Torill Ueland
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Trine V Lagerberg
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ingrid Melle
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
46
|
Sotelo-Ramírez CE, Valdés-Tovar M, Zaragoza-Hoyos JU, Ortiz-López L, Argueta J, Rosel-Vales M, Miranda-Labra RU, Camarena B. Molecular and Functional Analysis of TLR 1, 2 and 6 in Peripheral Blood Monocytes of Patients with Schizophrenia: A Pilot Study. Int J Mol Sci 2025; 26:926. [PMID: 39940697 PMCID: PMC11817014 DOI: 10.3390/ijms26030926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 02/16/2025] Open
Abstract
Schizophrenia (SZ) is a chronic disabling mental disorder with high heritability, and several immune-regulating genes have been implicated in its pathophysiology In this study, we investigated the expression of Toll-like receptors (TLRs) 1, 2, and 6 in peripheral blood monocytes from SZ patients and healthy control subjects (HCSs) in the Mexican population, focusing on specific SZ-associated gene variants. Gene expressions were assessed by qPCR, and protein expression was measured using flow cytometry. The secretory profiles of MALP2-stimulated monocytes were evaluated through immunoproteomic arrays. Our results indicate that patients with SZ carrying the rs4833093/TLR1 GG genotype exhibited significantly lower TLR1 gene expression compared to TT carriers. Notably, HCSs with the TT genotype showed markedly higher TLR1 protein expression, while all patients with SZ exhibited significantly reduced protein levels regardless of genotype. Furthermore, monocytes from patients with SZ displayed altered secretion profiles upon TLR stimulation, with significant elevations in IL-18, uPAR, angiopoietin-2, and serpin E1, alongside reductions in MCP-1, IL-17A, IL-24, MIF, and myeloperoxidase compared to HCSs. These findings suggest a dysfunctional TLR-mediated innate immune response in SZ.
Collapse
Affiliation(s)
- Carlo E. Sotelo-Ramírez
- Doctorado en Biología Experimental, Universidad Autónoma Metropolitana (UAM)-Iztapalapa, Mexico City 09340, Mexico;
- Departamento de Farmacogenética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| | - Marcela Valdés-Tovar
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (M.V.-T.); (L.O.-L.)
| | - Julio Uriel Zaragoza-Hoyos
- Departamento de Farmacogenética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| | - Leonardo Ortiz-López
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (M.V.-T.); (L.O.-L.)
| | - Jesús Argueta
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| | - Mauricio Rosel-Vales
- Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Roxana U. Miranda-Labra
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana (UAM)-Iztapalapa, Mexico City 09340, Mexico
| | - Beatriz Camarena
- Departamento de Farmacogenética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| |
Collapse
|
47
|
Surniyantoro HNE, Tetriana D, Yusuf D, Mailana W, Indriatama WM, Nasution KY, Purnami S, Rahajeng N, Lusiyanti Y, Kisnanto T. Association of -607C/A (rs1946518) and -137G/C (rs187238) polymorphisms and immune response in radiation-exposed workers. Int J Radiat Biol 2025; 101:351-357. [PMID: 39823224 DOI: 10.1080/09553002.2025.2452293] [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: 06/26/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE Interleukin-18, transforming growth factor-β, and superoxide dismutase are important cytokines and antioxidants in protecting the body from damage caused by radiation exposure through an immune response mechanism. Genetic polymorphisms -607 C/A and -137 G/C are thought to affect the IL-18 cytokine in carrying out its function as a biomarker to indicate adverse conditions due to radiation. The purposes of this study were to investigate the association between 607 C/A and -137 G/C SNPs on the concentrations of IL-18, and to measure TGF-β and SOD activity in radiation workers and control group. MATERIAL AND METHOD We enrolled 40 radiation workers and 40 non-radiation workers as a control group. We determined genotype distribution of -607 C/A and -137 G/C SNPs and their correlation with IL-18 concentration by using PCR-RFLP method. We also measured the IL-18, TGF-β concentration, and SOD activity by using Elisa assay. RESULTS AND CONCLUSION No relationship was found between -607 C/A and -137 G/C on IL-18 concentrations in all genotype groups, and no significant difference in IL-18 and TGF-β concentrations in the radiation worker and control groups. Significant differences were found only in lower SOD activity in radiation workers compared to controls. The -607 C/A and -137 G/C did not significantly correlate with IL-18 cytokine production in all genotypes. There was no significant difference between IL-18 and TGF-β concentrations in the radiation worker and control groups. However, there was a very significant decrease in the SOD activity of the radiation workers by 3.31 times compared to the controls.
Collapse
Affiliation(s)
- Harry Nugroho Eko Surniyantoro
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Serpong, Indonesia
| | - Devita Tetriana
- Research Center for Safety, Metrology, and Nuclear Quality Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Darlina Yusuf
- Research Center for Safety, Metrology, and Nuclear Quality Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Wiwin Mailana
- Research Center for Safety, Metrology, and Nuclear Quality Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Wijaya Murti Indriatama
- Research Center for Radiation Process Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Khairul Yusuf Nasution
- Research Center for Radiation Process Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Sofiati Purnami
- Research Center for Safety, Metrology, and Nuclear Quality Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Nastiti Rahajeng
- Research Center for Safety, Metrology, and Nuclear Quality Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Yanti Lusiyanti
- Research Center for Safety, Metrology, and Nuclear Quality Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Teja Kisnanto
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Serpong, Indonesia
| |
Collapse
|
48
|
Khalil ZHA, Osman TA. Multifocal osteochondromatous proliferation and paraneoplastic hematologic dyscrasia in the context of latent Epstein-Barr virus reactivation: a case of oncologic and infectious pathophysiology. Skeletal Radiol 2025:10.1007/s00256-025-04872-y. [PMID: 39821684 DOI: 10.1007/s00256-025-04872-y] [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: 09/19/2024] [Revised: 11/28/2024] [Accepted: 01/04/2025] [Indexed: 01/19/2025]
Abstract
This case report describes a 15-year-old male with multifocal osteochondromatous proliferation and paraneoplastic hematologic dyscrasia, linked to latent Epstein-Barr virus reactivation. Radiographic and advanced imaging revealed widespread skeletal lesions consistent with osteochondromatosis. Hematologic evaluation indicated pancytopenia with dysplastic megakaryocytes and marrow infiltration. Immunohistochemical staining confirmed latent Epstein-Barr virus infection, suggesting its role in the pathogenesis of both the osteochondromatous and hematologic abnormalities. This case highlights the correlation between Epstein-Barr virus reactivation, bone proliferation, and paraneoplastic hematologic processes, which we believe has not yet been reported in the literature, emphasizing the need for a comprehensive diagnostic approach.
Collapse
Affiliation(s)
- Zeyad Hossam Atta Khalil
- College of Medicine/Radiology Department, October 6, University, 217G Pyramid Gardens, Cairo, Egypt.
| | - Taha Ali Osman
- College of Medicine/Radiology Department, October 6, University, 217G Pyramid Gardens, Cairo, Egypt
| |
Collapse
|
49
|
Vijayan S, Margesan T. Hormonal Imbalance in Obesity and Arthritis: Points of Contact. Curr Rheumatol Rev 2025; 21:182-193. [PMID: 38623986 DOI: 10.2174/0115733971293288240313090945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 04/17/2024]
Abstract
Obesity is a growing global health crisis intricately connected to various chronic conditions, including arthritis. This paper explores the intricate web of hormonal changes in the context of obesity and their profound influence on the development and progression of arthritis. Hormones, such as leptin, insulin, cortisol, and estrogen, all altered in obesity, play pivotal roles in inflammation, cartilage degradation, mechanical stress, and pain associated with obesity-related arthritis. Additionally, the mechanical stress placed on weight-bearing joints by excess body weight accelerates joint wear and tear, contributing to arthritis. Genetic factors, shared biomarkers, and pathways further link these conditions. Recognizing these connections is vital for healthcare professionals and individuals facing the challenges of obesity and arthritis, offering insights into strategies for prevention, management, and intervention. This comprehensive understanding of the complex interplay between hormonal changes, obesity, and arthritis sheds light on multifaceted mechanisms underlying this intricate relationship.
Collapse
Affiliation(s)
- Sukanya Vijayan
- Department of Pharmacognosy, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Thirumal Margesan
- Department of Pharmacognosy, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| |
Collapse
|
50
|
Huyen TT, Phuc NH, Lan PT, My LH, Vinh NTH, Son DA, Doanh LH. Serum Concentrations of IL-18, IL-21, IL-22, IL-23, IL-27, and IL-31 in Patients With Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis and Their Correlation With Disease Severity. Med Arch 2025; 79:9-14. [PMID: 40322299 PMCID: PMC12045582 DOI: 10.5455/medarh.2025.79.9-14] [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: 02/15/2025] [Accepted: 03/20/2025] [Indexed: 05/07/2025] Open
Abstract
Background Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are life-threatening conditions marked by extensive epidermal necrolysis and skin sloughing. Objective The study aimed to assess the serum levels of specific proinflammatory interleukins (IL-18, -21, 22, -23, -27, and -31) and their relationship with the severity of SJS/TEN within the Vietnamese population. Methods This descriptive cross-sectional study was conducted from 2018 to 2020. Serum levels of IL-18, -21, -22, -23, -27, and -31 were measured using the fluorescence covalent microbead immunosorbent assay. Results The study included 61 patients (29 males and 32 females; 21 with SJS and 40 with TEN), with a median age of 51 years (interquartile range: 37-58), and 20 healthy controls. The median lesional area covered 45% of the body surface area (interquartile range: 8-70%). The most frequently identified medications were traditional medicine (19 patients; 31.15%), allopurinol (9 patients; 14.75%), and carbamazepine (8 patients; 13.11%). In the TEN group, the serum level of IL-18 was significantly elevated compared to the healthy control group. A correlation was found between serum levels of IL-18 and IL-27 and the lesional area in SJS/TEN patients, as well as between serum levels of IL-18 and IL-31 and the lesional area in TEN patients. Conclusion Serum levels of IL-18 were increased in TEN group. Additionally, serum concentrations of IL-18, IL-27, and IL-31 were associated with disease severity as indicated by the lesional area. These interleukins may play an important role in the pathogenesis of SJS/TEN.
Collapse
Affiliation(s)
- Tran Thi Huyen
- Department of Dermatology and Venereology, Hanoi Medical University, Hanoi, Vietnam
- Department of Outpatient, National Hospital of Dermatology and Venereology, Hanoi, Vietnam
| | | | - Pham Thi Lan
- Department of Dermatology and Venereology, Hanoi Medical University, Hanoi, Vietnam
- Department of Outpatient, National Hospital of Dermatology and Venereology, Hanoi, Vietnam
| | - Le Huyen My
- Department of Outpatient, National Hospital of Dermatology and Venereology, Hanoi, Vietnam
| | - Nguyen Thi Ha Vinh
- Department of Dermatology and Venereology, Hanoi Medical University, Hanoi, Vietnam
- Department of Outpatient, National Hospital of Dermatology and Venereology, Hanoi, Vietnam
| | - Dao Anh Son
- Department of Health Finance and Health Technology Assessment, Health Strategy and Policy Institute
| | - Le Huu Doanh
- Department of Dermatology and Venereology, Hanoi Medical University, Hanoi, Vietnam
- Department of Outpatient, National Hospital of Dermatology and Venereology, Hanoi, Vietnam
| |
Collapse
|