Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
Int Immunopharmacol ; 113(Pt A): 109424, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36461589

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease of unknown cause and characterized by excessive proliferation of fibroblasts and the irregular remodeling of extracellular matrix (ECM), which ultimately cause the severe distortion of the alveolar architecture. The median survival of IPF patients is 2-5 years. IPF patients are predominantly infiltrated by M2 macrophages during the course of disease development and progression. Predominantly accumulation of M2 macrophages accelerates fibrosis progression by secreting multiple cytokines that promote fibroblast to myofibroblast transition. In the process of M2 macrophage polarization, JAK2/STAT3 signaling plays a key role, thus, targeting activated macrophages to inhibit the pro-fibrotic phenotype is considered as an approach to the potential treatment of IPF. Tacrolimus is a macrolide antibiotic that as a specific inhibitor of T-lymphocyte function and has been used widely as an immunosuppressant in human organ transplantation. In this study we explored the potential effect and mechanism of tacrolimus on pulmonary fibrosis in vivo and vitro. Here, we found that tacrolimus is capable of suppressing M2 macrophages polarization by inhibiting pro-fibrotic factors secreted by M2 macrophages. This effect further alleviates M2-induced myofibroblast activation, thus resulting in a decline of collagen deposition, pro-fibrotic cytokines secretion, recovering of lung function, ultimately relieving the progression of fibrosis in vivo. Mechanistically, we found that tacrolimus can inhibit the activation of JAK2/STAT3 signaling by targeting JAK2. Our findings indicate a potential anti-fibrotic effect of tacrolimus by regulating macrophage polarization and might be meaningful in clinical settings.


Subject(s)
Bleomycin , Idiopathic Pulmonary Fibrosis , Macrophages , Tacrolimus , Humans , Bleomycin/adverse effects , Cytokines , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/immunology , Janus Kinase 2/immunology , Macrophages/drug effects , Macrophages/immunology , STAT3 Transcription Factor/immunology , Tacrolimus/pharmacology , Tacrolimus/therapeutic use
2.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163413

ABSTRACT

Neutrophils are an essential component of the innate immune response, but their prolonged activation can lead to chronic inflammation. Consequently, neutrophil homeostasis is tightly regulated through balance between granulopoiesis and clearance of dying cells. The bone marrow is both a site of neutrophil production and the place they return to and die. Myeloproliferative neoplasms (MPN) are clonal hematopoietic disorders characterized by the mutations in three types of molecular markers, with emphasis on Janus kinase 2 gene mutation (JAK2V617F). The MPN bone marrow stem cell niche is a site of chronic inflammation, with commonly increased cells of myeloid lineage, including neutrophils. The MPN neutrophils are characterized by the upregulation of JAK target genes. Additionally, MPN neutrophils display malignant nature, they are in a state of activation, and with deregulated apoptotic machinery. In other words, neutrophils deserve to be placed in the midst of major events in MPN. Our crucial interest in this review is better understanding of how neutrophils die in MPN mirrored by defects in apoptosis and to what possible extent they can contribute to MPN pathophysiology. We tend to expect that reduced neutrophil apoptosis will establish a pathogenic link to chronic inflammation in MPN.


Subject(s)
Hematologic Neoplasms/immunology , Immunity, Innate , Myeloproliferative Disorders/immunology , Neutrophils/immunology , Amino Acid Substitution , Animals , Chronic Disease , Hematologic Neoplasms/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Mutation, Missense , Myeloproliferative Disorders/genetics
3.
Biomed Environ Sci ; 35(2): 95-106, 2022 Feb 20.
Article in English | MEDLINE | ID: mdl-35197174

ABSTRACT

OBJECTIVE: This study aimed to investigate the effects of caprylic acid (C8:0) on lipid metabolism and inflammation, and examine the mechanisms underlying these effects in mice and cells. METHODS: Fifty-six 6-week-old male C57BL/6J mice were randomly allocated to four groups fed a high-fat diet (HFD) without or with 2% C8:0, palmitic acid (C16:0) or eicosapentaenoic acid (EPA). RAW246.7 cells were randomly divided into five groups: normal, lipopolysaccharide (LPS), LPS+C8:0, LPS+EPA and LPS+cAMP. The serum lipid profiles, inflammatory biomolecules, and ABCA1 and JAK2/STAT3 mRNA and protein expression were measured. RESULTS: C8:0 decreased TC and LDL-C, and increased the HDL-C/LDL-C ratio after injection of LPS. Without LPS, it decreased TC in mice ( P < 0.05). Moreover, C8:0 decreased the inflammatory response after LPS treatment in both mice and cells ( P < 0.05). Mechanistic investigations in C57BL/6J mouse aortas after injection of LPS indicated that C8:0 resulted in higher ABCA1 and JAK2/STAT3 expression than that with HFD, C16:0 and EPA, and resulted in lower TNF-α, NF-κB mRNA expression than that with HFD ( P < 0.05). In RAW 264.7 cells, C8:0 resulted in lower expression of pNF-κBP65 than that in the LPS group, and higher protein expression of ABCA1, p-JAK2 and p-STAT3 than that in the LPS and LPS+cAMP groups ( P < 0.05). CONCLUSION: Our studies demonstrated that C8:0 may play an important role in lipid metabolism and the inflammatory response, and the mechanism may be associated with ABCA1 and the p-JAK2/p-STAT3 signaling pathway.


Subject(s)
ATP Binding Cassette Transporter 1/immunology , Caprylates/administration & dosage , Inflammation/drug therapy , Janus Kinase 2/immunology , Lipid Metabolism/drug effects , Macrophages/drug effects , STAT3 Transcription Factor/immunology , ATP Binding Cassette Transporter 1/genetics , Animals , Caprylates/chemistry , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Janus Kinase 2/genetics , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , STAT3 Transcription Factor/genetics , Signal Transduction
4.
Article in English | WPRIM (Western Pacific) | ID: wpr-927639

ABSTRACT

OBJECTIVE@#This study aimed to investigate the effects of caprylic acid (C8:0) on lipid metabolism and inflammation, and examine the mechanisms underlying these effects in mice and cells.@*METHODS@#Fifty-six 6-week-old male C57BL/6J mice were randomly allocated to four groups fed a high-fat diet (HFD) without or with 2% C8:0, palmitic acid (C16:0) or eicosapentaenoic acid (EPA). RAW246.7 cells were randomly divided into five groups: normal, lipopolysaccharide (LPS), LPS+C8:0, LPS+EPA and LPS+cAMP. The serum lipid profiles, inflammatory biomolecules, and ABCA1 and JAK2/STAT3 mRNA and protein expression were measured.@*RESULTS@#C8:0 decreased TC and LDL-C, and increased the HDL-C/LDL-C ratio after injection of LPS. Without LPS, it decreased TC in mice ( P < 0.05). Moreover, C8:0 decreased the inflammatory response after LPS treatment in both mice and cells ( P < 0.05). Mechanistic investigations in C57BL/6J mouse aortas after injection of LPS indicated that C8:0 resulted in higher ABCA1 and JAK2/STAT3 expression than that with HFD, C16:0 and EPA, and resulted in lower TNF-α, NF-κB mRNA expression than that with HFD ( P < 0.05). In RAW 264.7 cells, C8:0 resulted in lower expression of pNF-κBP65 than that in the LPS group, and higher protein expression of ABCA1, p-JAK2 and p-STAT3 than that in the LPS and LPS+cAMP groups ( P < 0.05).@*CONCLUSION@#Our studies demonstrated that C8:0 may play an important role in lipid metabolism and the inflammatory response, and the mechanism may be associated with ABCA1 and the p-JAK2/p-STAT3 signaling pathway.


Subject(s)
Animals , Humans , Male , Mice , ATP Binding Cassette Transporter 1/immunology , Caprylates/chemistry , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Inflammation/metabolism , Janus Kinase 2/immunology , Lipid Metabolism/drug effects , Macrophages/immunology , Mice, Inbred C57BL , STAT3 Transcription Factor/immunology , Signal Transduction
5.
Front Immunol ; 12: 734652, 2021.
Article in English | MEDLINE | ID: mdl-34867954

ABSTRACT

Microbial challenges, such as widespread bacterial infection in sepsis, induce endotoxin tolerance, a state of hyporesponsiveness to subsequent infections. The participation of DNA methylation in this process is poorly known. In this study, we perform integrated analysis of DNA methylation and transcriptional changes following in vitro exposure to gram-negative bacterial lipopolysaccharide, together with analysis of ex vivo monocytes from septic patients. We identify TET2-mediated demethylation and transcriptional activation of inflammation-related genes that is specific to toll-like receptor stimulation. Changes also involve phosphorylation of STAT1, STAT3 and STAT5, elements of the JAK2 pathway. JAK2 pathway inhibition impairs the activation of tolerized genes on the first encounter with lipopolysaccharide. We then confirm the implication of the JAK2-STAT pathway in the aberrant DNA methylome of patients with sepsis caused by gram-negative bacteria. Finally, JAK2 inhibition in monocytes partially recapitulates the expression changes produced in the immunosuppressive cellular state acquired by monocytes from gram-negative sepsis, as described by single cell-RNA-sequencing. Our study evidences both the crucial role the JAK2-STAT pathway in epigenetic regulation and initial response of the tolerized genes to gram-negative bacterial endotoxins and provides a pharmacological target to prevent exacerbated responses.


Subject(s)
Endotoxin Tolerance/genetics , Gram-Negative Bacteria/immunology , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Monocytes/immunology , Monocytes/microbiology , Sepsis/genetics , Sepsis/immunology , Case-Control Studies , DNA Methylation/genetics , DNA Methylation/immunology , Endotoxin Tolerance/drug effects , Endotoxin Tolerance/immunology , Endotoxins/toxicity , Epigenesis, Genetic , Female , Gram-Negative Bacterial Infections/microbiology , Humans , In Vitro Techniques , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lipopolysaccharides/toxicity , Male , Monocytes/drug effects , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Sepsis/microbiology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology
6.
Cell Rep Med ; 2(10): 100411, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34755131

ABSTRACT

Neoadjuvant PD-1 blockade may be efficacious in some individuals with high-risk, resectable oral cavity head and neck cancer. To explore correlates of response patterns to neoadjuvant nivolumab treatment and post-surgical recurrences, we analyzed longitudinal tumor and blood samples in a cohort of 12 individuals displaying 33% responsiveness. Pretreatment tumor-based detection of FLT4 mutations and PTEN signature enrichment favors response, and high tumor mutational burden improves recurrence-free survival. In contrast, preexisting and/or acquired mutations (in CDKN2A, YAP1, or JAK2) correlate with innate resistance and/or tumor recurrence. Immunologically, tumor response after therapy entails T cell receptor repertoire diversification in peripheral blood and intratumoral expansion of preexisting T cell clones. A high ratio of regulatory T to T helper 17 cells in pretreatment blood predicts low T cell receptor repertoire diversity in pretreatment blood, a low cytolytic T cell signature in pretreatment tumors, and innate resistance. Our study provides a molecular framework to advance neoadjuvant anti-PD-1 therapy for individuals with resectable head and neck cancer.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Mouth Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/surgery , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/immunology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Inhibitors/therapeutic use , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Mouth Neoplasms/genetics , Mouth Neoplasms/immunology , Mouth Neoplasms/surgery , Mutation , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/surgery , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Survival Analysis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/pathology , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-3/immunology , YAP-Signaling Proteins/genetics , YAP-Signaling Proteins/immunology
7.
J Clin Invest ; 131(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34730109

ABSTRACT

Despite the curative potential of hematopoietic stem cell transplantation (HSCT), conditioning-associated toxicities preclude broader clinical application. Antibody-drug conjugates (ADCs) provide an attractive approach to HSCT conditioning that minimizes toxicity while retaining efficacy. Initial studies of ADC conditioning have largely focused on syngeneic HSCT. However, to treat acute leukemias or induce tolerance for solid organ transplantation, this approach must be expanded to allogeneic HSCT (allo-HSCT). Using murine allo-HSCT models, we show that pharmacologic Janus kinase 1/2 (JAK1/2) inhibition combined with CD45- or cKit-targeted ADCs enables robust multilineage alloengraftment. Strikingly, myeloid lineage donor chimerism exceeding 99% was achievable in fully MHC-mismatched HSCT using this approach. Mechanistic studies using the JAK1/2 inhibitor baricitinib revealed marked impairment of T and NK cell survival, proliferation, and effector function. NK cells were exquisitely sensitive to JAK1/2 inhibition due to interference with IL-15 signaling. Unlike irradiated mice, ADC-conditioned mice did not develop pathogenic graft-versus-host alloreactivity when challenged with mismatched T cells. Finally, the combination of ADCs and baricitinib balanced graft-versus-host disease and graft-versus-leukemia responses in delayed donor lymphocyte infusion models. Our allo-HSCT conditioning strategy exemplifies the promise of immunotherapy to improve the safety of HSCT for treating hematologic diseases.


Subject(s)
Azetidines/pharmacology , Hematopoietic Stem Cell Transplantation , Immunoconjugates/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyrazoles/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Allografts , Animals , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/drug effects , Graft vs Leukemia Effect/genetics , Graft vs Leukemia Effect/immunology , Interleukin-15/genetics , Interleukin-15/immunology , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology
8.
Front Immunol ; 12: 704526, 2021.
Article in English | MEDLINE | ID: mdl-34497607

ABSTRACT

Objective: Baricitinib, a selective inhibitor for janus kinase (JAK) 1 and JAK2, is approved for use in rheumatoid arthritis. Systemic lupus erythematosus (SLE) is recently regarded as a potential candidate targeted by JAK inhibitors because of the relationship between its pathogenesis and JAK/signal transducer and activator of transcription (STAT) pathway-mediated cytokines such as type I interferons. The objective of this study was to determine whether baricitinib could effectively ameliorate SLE using a murine model. Methods: To investigate effects of baricitinib on various autoimmune features, especially renal involvements in SLE, eight-week-old MRL/Mp-Faslpr (MRL/lpr) mice were used as a lupus-prone animal model and treated with baricitinib for eight weeks. Immortalized podocytes and primary podocytes and B cells isolated from C57BL/6 mice were used to determine the in vitro efficacy of baricitinib. Results: Baricitinib remarkably suppressed lupus-like phenotypes of MRL/lpr mice, such as splenomegaly, lymphadenopathy, proteinuria, and systemic autoimmunity including circulating autoantibodies and pro-inflammatory cytokines. It also modulated immune cell populations and effectively ameliorated renal inflammation, leading to the recovery of the expression of structural proteins in podocytes. According to in vitro experiments, baricitinib treatment could mitigate B cell differentiation and restore disrupted cytoskeletal structures of podocytes under inflammatory stimulation by blocking the JAK/STAT pathway. Conclusions: The present study demonstrated that baricitinib could effectively attenuate autoimmune features including renal inflammation of lupus-prone mice by suppressing aberrant B cell activation and podocyte abnormalities. Thus, baricitinib as a selective JAK inhibitor could be a promising therapeutic candidate in the treatment of SLE.


Subject(s)
Azetidines/pharmacology , Lupus Erythematosus, Systemic , Podocytes , Purines/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Disease Models, Animal , Female , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred MRL lpr , Podocytes/immunology , Podocytes/pathology , Signal Transduction/drug effects
9.
Biochem Pharmacol ; 192: 114690, 2021 10.
Article in English | MEDLINE | ID: mdl-34274356

ABSTRACT

BACKGROUND: Eosinophilic asthma is increasingly recognized as one of the most severe and difficult-to-treat asthma subtypes. The JAK/STAT pathway is the principal signaling mechanism for a variety of cytokines and growth factors involved in asthma. However, the direct effect of JAK inhibitors on eosinophil effector function has not been addressed thus far. OBJECTIVE: Here we compared the effects of the JAK1/2 inhibitor baricitinib and the JAK3 inhibitor tofacitinib on eosinophil effector function in vitro and in vivo. METHODS: Differentiation of murine bone marrow-derived eosinophils. Migratory responsiveness, respiratory burst, phagocytosis and apoptosis of human peripheral blood eosinophils were assessed in vitro. In vivo effects were investigated in a mouse model of acute house dust mite-induced airway inflammation in BALB/c mice. RESULTS: Baricitinib more potently induced apoptosis and inhibited eosinophil chemotaxis and respiratory burst, while baricitinib and tofacitinib similarly affected eosinophil differentiation and phagocytosis. Of the JAK inhibitors, oral application of baricitinib more potently prevented lung eosinophilia in mice following allergen challenge. However, both JAK inhibitors neither affected airway resistance nor compliance. CONCLUSION: Our data suggest that the JAK1/2 inhibitor baricitinib is even more potent than the JAK3 inhibitor tofacitinib in suppressing eosinophil effector function. Thus, targeting the JAK1/2 pathway represents a promising therapeutic strategy for eosinophilic inflammation as observed in severe eosinophilic asthma.


Subject(s)
Azetidines/therapeutic use , Eosinophilia/drug therapy , Eosinophils/drug effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/therapeutic use , Purines/therapeutic use , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Adult , Animals , Azetidines/pharmacology , Cells, Cultured , Eosinophilia/chemically induced , Eosinophilia/immunology , Eosinophils/physiology , Female , Humans , Janus Kinase 1/immunology , Janus Kinase 2/immunology , Janus Kinase Inhibitors/pharmacology , Male , Mice , Mice, Inbred BALB C , Purines/pharmacology , Pyrazoles/pharmacology , Pyroglyphidae/immunology , Sulfonamides/pharmacology , Young Adult
10.
Inflammopharmacology ; 29(4): 1101-1109, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34218389

ABSTRACT

There are accumulating reports regarding poor response to common antidepressant therapy. Antidepressant resistance is often linked to inflammatory system activation and patients displaying inflammation prior to the treatment are less responsive to antidepressants. We hypothesized that the inefficacy of antidepressant therapy in some patients may be attributable to the drugs' inflammatory mode of action, which has been overlooked because of their substantial therapeutic benefit. Bupropion is a commonly prescribed antidepressant that is often used to treat seasonal affective disorders as well. Nevertheless, research suggests that bupropion causes inflammation and worsens depressive symptoms. Therefore, we investigated the impact of bupropion on cytokines of innate and adaptive immunity, as well as immune signaling pathways. We treated lipopolysaccharide (LPS)-stimulated human peripheral blood mononuclear cells (PBMCs) with different doses of bupropion. Pro-/anti-inflammatory cytokines [tumor necrosis factor alpha (TNFα), interleukin-1ß (IL-1ß), IL-17, and IL-10] were assessed at both transcriptional and translational levels as well as the involvement of JAK2 /STAT3, TLR2, and TLR4 signaling in this process. Bupropion reduced IL-17A, TNFα, and IL-1ß protein levels in the cultures. Nonetheless, bupropion increased IL-1ß (P < 0.0001), TNFα (P < 0.0001), and IL-17A (P < 0.05) mRNA levels. Treatment enhanced both IL-10 concentration (P < 0.0001) and gene expression (P < 0.0001). TLR2 (P < 0.0001), TLR4 (P < 0.0001), JAK2 (P < 0.0001), and STAT3 (P < 0.0001) gene expression also rose in response to bupropion. The findings imply that bupropion, particularly 50 µM and 100 µM, has pro-inflammatory effects and should be co-administered with anti-inflammatory medications, at least in patients with inflammatory conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bupropion/pharmacology , Janus Kinase 2/biosynthesis , STAT3 Transcription Factor/biosynthesis , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 4/biosynthesis , Adult , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Janus Kinase 2/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , STAT3 Transcription Factor/agonists , STAT3 Transcription Factor/immunology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Young Adult
11.
Blood ; 138(12): 1034-1039, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34232994

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is an inflammatory disorder in which numerous cytokines are elevated, though interferon-γ (IFN-γ) is central to disease pathogenesis and a key therapeutic target. Experimental and early clinical reports have shown that ruxolitinib, a small molecule inhibitor of Janus kinases (JAKs), which are essential for cytokine signaling, may be therapeutic in HLH. In contrast, we found that intermittently administered ruxolitinib at various dose levels failed to prevent HLH development or treat established murine HLH. High doses of ruxolitinib blocked IFN-γ signaling only transiently after administration, consistent with human pharmacokinetics, and only continuously administered drug could prevent HLH development or treat established HLH. Continuously administered ruxolitinib was therapeutic in only a narrow dose range and intermittently dosed ruxolitinib worsened survival and decreased bone marrow cellularity of animals concurrently treated with anti-IFN-γ antibody, indicating a narrow therapeutic window and potential toxicity. Because JAK2 is essential for hematopoietic cytokine signaling, we also tested a JAK1-selective inhibitor and observed therapeutic benefit without apparent toxicity, though it did not improve survival when combined with anti-IFN-γ. We conclude that continuous blockade of IFN-γ signaling is necessary for optimal control of HLH and that JAK2 inhibition may be toxic in this disorder.


Subject(s)
Interferon-gamma/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/immunology , Lymphohistiocytosis, Hemophagocytic , Nitriles/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Animals , Disease Models, Animal , Janus Kinase 2/genetics , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/immunology , Mice , Signal Transduction/immunology
12.
Hepatology ; 74(3): 1395-1410, 2021 09.
Article in English | MEDLINE | ID: mdl-33738839

ABSTRACT

BACKGROUND AND AIMS: Recent development of multiple treatments for human hepatocellular carcinoma (HCC) has allowed for the selection of combination therapy to enhance the effectiveness of monotherapy. Optimal selection of therapies is based on both HCC and its microenvironment. Therefore, it is critical to develop and validate preclinical animal models for testing clinical therapeutic solutions. APPROACH AND RESULTS: We established cell line-based or patient-derived xenograft-based humanized-immune-system mouse models with subcutaneous and orthotopic HCC. Mice were injected with human-specific antibodies (Abs) to deplete human immune cells. We analyzed the transcription profiles of HCC cells and human immune cells by using real-time PCR and RNA sequencing. The protein level of HCC tumor cells/tissues or human immune cells was determined by using flow cytometry, western blotting, and immunohistochemistry. The HCC tumor size was measured after single, dual-combination, and triple-combination treatment using N-(1',2-Dihydroxy-1,2'-binaphthalen-4'-yl)-4-methoxybenzenesulfonamide (C188-9), bevacizumab, and pembrolizumab. In this study, human immune cells in the tumor microenvironment were strongly selected and modulated by HCC, which promoted the activation of the IL-6/Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in tumor cells and led to augmented HCC proliferation and angiogenesis by releasing angiogenic cytokines in humanized-immune-system mice with HCC. In particular, intratumor human cluster of differentiation-positive (hCD14+ ) cells could produce IL-33 through damage-associated molecular pattern/Toll-like receptor 4/activator protein 1, which up-regulated IL-6 in other intratumor immune cells and activated the JAK2/STAT3 pathway in HCC. Specific knockdown of the CD14 gene in human monocytes could impair IL-33 production induced by cell lysates. Subsequently, we evaluated the in vivo anti-HCC effect of C188-9, bevacizumab, and pembrolizumab. The results showed that the anti-HCC effect of triple-combination therapy was superior to that of single or dual treatments. CONCLUSIONS: Humanized-immune-system HCC mouse models are suitable for identifying targets from cancer and immune components and for testing combinational therapies.


Subject(s)
Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neovascularization, Pathologic/immunology , Tumor Microenvironment/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Bevacizumab/pharmacology , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Interleukin-6/immunology , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lipopolysaccharide Receptors/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Mice , Naphthols/pharmacology , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction , Sulfonamides/pharmacology , Transcriptome , Xenograft Model Antitumor Assays
13.
Int J Mol Sci ; 22(4)2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33672997

ABSTRACT

The Philadelphia-negative myeloproliferative neoplasms (MPNs) are malignancies of the hematopoietic stem cell (HSC) arising as a consequence of clonal proliferation driven by somatically acquired driver mutations in discrete genes (JAK2, CALR, MPL). In recent years, along with the advances in molecular characterization, the role of immune dysregulation has been achieving increasing relevance in the pathogenesis and evolution of MPNs. In particular, a growing number of studies have shown that MPNs are often associated with detrimental cytokine milieu, expansion of the monocyte/macrophage compartment and myeloid-derived suppressor cells, as well as altered functions of T cells, dendritic cells and NK cells. Moreover, akin to solid tumors and other hematological malignancies, MPNs are able to evade T cell immune surveillance by engaging the PD-1/PD-L1 axis, whose pharmacological blockade with checkpoint inhibitors can successfully restore effective antitumor responses. A further interesting cue is provided by the recent discovery of the high immunogenic potential of JAK2V617F and CALR exon 9 mutations, that could be harnessed as intriguing targets for innovative adoptive immunotherapies. This review focuses on the recent insights in the immunological dysfunctions contributing to the pathogenesis of MPNs and outlines the potential impact of related immunotherapeutic approaches.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunotherapy/methods , Inflammation/immunology , Myeloproliferative Disorders/therapy , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Calreticulin/genetics , Calreticulin/immunology , Calreticulin/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Inflammation/genetics , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Janus Kinase 2/metabolism , Mutation/immunology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Philadelphia Chromosome , T-Lymphocytes/metabolism , Tumor Microenvironment/genetics
14.
Food Funct ; 11(10): 8987-8995, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33001081

ABSTRACT

Obesity is an increasingly severe global health problem, leading to chronic inflammation and metabolic disorders in both peripheral tissues and the central nervous system. Matcha is a powdered green tea, and it is very popular in recent years as a beverage and food additive. Matcha green tea has been reported to have outstanding potential in regulating obesity-related metabolic syndrome. However, there are few studies on the regulation mechanism of matcha green tea on the central nervous system. In this study, we established a high-fat diet-induced obese mouse model. The results showed that dietary supplementation with matcha could effectively inhibit the weight gain, fat accumulation, glycemia and lipidemia increase, and excessive activation of microglia in the arcuate nucleus of the hypothalamus. Furthermore, we used different concentrations (100%, 80%, 60%, 40%, and 20%, v/v) of ethanol solution to prepare matcha ethanol extracts, and investigated their effects on palmitic acid-induced inflammation of microglial BV-2 cells. The results showed that matcha ethanol extracts could significantly reduce the release of inflammatory cytokines and the expression and phosphorylation of JAK2 and STAT3.


Subject(s)
Camellia sinensis/chemistry , Hypothalamus/drug effects , Janus Kinase 2/immunology , Obesity/drug therapy , Obesity/immunology , Plant Extracts/administration & dosage , STAT3 Transcription Factor/immunology , Animals , Antioxidants/administration & dosage , Humans , Hypothalamus/immunology , Janus Kinase 2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Tea/chemistry
15.
Exp Hematol ; 92: 75-88.e10, 2020 12.
Article in English | MEDLINE | ID: mdl-33017633

ABSTRACT

Human leukocyte antigen class I (HLA-I) genotype has been found to influence cancer development through the presentation of mutational neoepitopes. However, our understanding of its effect on the development of myeloproliferative neoplasms (MPNs) remains limited. We aimed to elucidate the putative protective role of HLA-I alleles in the development of JAK2 V617F-driven MPNs using a population genetics approach. The variability of the HLA-I genotype had no effect on the presence of JAK2 V617F mutation. However, three alleles were found to be inversely correlated with the presence of JAK2 V617F mutation: HLA-A*02:01 (p = 0.036), HLA-B*35:01 (p = 0.017), and HLA-C*15:02 (p = 0.033). The HLA-B*35:01 allele was predicted to bind to a 9-mer peptide derived from JAK2 V617F mutant protein. Gene expression analysis revealed a lower expression of HLA-A and -B in MPN CD34+ cells compared with normal CD34+ cells, which was modulated by ruxolitinib and interferon-α treatment. In summary, we provide robust evidence that specific HLA-I molecules restrict JAK2 V617F-driven oncogenesis. JAK2 V617F+ stem cells evade immune surveillance through downregulation of the HLA-I expression. Therefore, the presence of specific HLA-I alleles might be a predictive marker for response to certain immunotherapies upregulating HLA-I expression. Finally, our findings have implications in the development of mutational neoepitope-based vaccines in MPNs.


Subject(s)
Alleles , Carcinogenesis , Hematologic Neoplasms , Histocompatibility Antigens Class I , Janus Kinase 2 , Mutation, Missense , Myeloproliferative Disorders , Amino Acid Substitution , Carcinogenesis/genetics , Carcinogenesis/immunology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology
16.
Int Immunopharmacol ; 88: 106891, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32853927

ABSTRACT

BACKGROUND: The therapeutic approaches guided toward microRNAs (miRNAs) have been extensively explored in lupus nephritis (LN), but the precise position of miR-10a-3p posted in disease is not translated thoroughly. Therein, this work pivoting on miR-10a-3p was launched with the involvement of regenerating islet-derived 3 α (REG3A). METHODS: Peripheral blood samples from LN patients and healthy controls (n = 132) were collected. miR-10a-3p and REG3A expression in peripheral blood mononuclear cells were tested. Mice were injected with miR-10a-3p agomir, miR-10a-3p antagomir and/or REG3A low expression vector for presentation of their roles in renal function, T helper cell 17 (Th17)/regulatory cell (Treg) balance, renal pathological damage, JAK2/STAT3 pathway activation and renal injury in LN. The relation between miR-10a-3p and REG3A was tested. RESULTS: MiR-10a-3p was down-regulated while REG3A was up-regulated in LN. Restoring miR-10a-3p or silencing REG3A decreased Th17/Treg ratio in CD4+ T cells, inhibited JAK2/STAT3 pathway activation, ameliorated renal function, improved renal pathological damage and alleviated renal injury in LN. REG3A depletion negated the effects of down-regulated miR-10a-3p on LN. MiR-10a-3p targeted REG3A. CONCLUSION: The work elucidates that miR-10a-3p restoration decreases Th17/Treg ratio and attenuates renal injury in LN via inhibiting REG3A and the activation of JAK2/STAT3 pathway, which renews the therapeutic reference for LN management.


Subject(s)
Lupus Nephritis/immunology , MicroRNAs , Pancreatitis-Associated Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adult , Animals , Female , Humans , Janus Kinase 2/immunology , Kidney/immunology , Lupus Nephritis/blood , Mice, Inbred C57BL , Middle Aged , Pancreatitis-Associated Proteins/genetics , STAT3 Transcription Factor/immunology , Spleen/cytology
17.
Clin Immunol ; 218: 108517, 2020 09.
Article in English | MEDLINE | ID: mdl-32585295

ABSTRACT

Approximately 15% of patients with coronavirus disease 2019 (COVID-19) experience severe disease, and 5% progress to critical stage that can result in rapid death. No vaccines or antiviral treatments have yet proven effective against COVID-19. Patients with severe COVID-19 experience elevated plasma levels of pro-inflammatory cytokines, which can result in cytokine storm, followed by massive immune cell infiltration into the lungs leading to alveolar damage, decreased lung function, and rapid progression to death. As many of the elevated cytokines signal through Janus kinase (JAK)1/JAK2, inhibition of these pathways with ruxolitinib has the potential to mitigate the COVID-19-associated cytokine storm and reduce mortality. This is supported by preclinical and clinical data from other diseases with hyperinflammatory states, where ruxolitinib has been shown to reduce cytokine levels and improve outcomes. The urgent need for treatments for patients with severe disease support expedited investigation of ruxolitinib for patients with COVID-19.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/prevention & control , Cytokines/antagonists & inhibitors , Pneumonia, Viral/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Severe Acute Respiratory Syndrome/prevention & control , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Betacoronavirus/immunology , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Cytokines/genetics , Cytokines/immunology , Drug Dosage Calculations , Gene Expression Regulation , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Nitriles , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyrimidines , SARS-CoV-2 , Severe Acute Respiratory Syndrome/complications , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/virology , Severity of Illness Index , Signal Transduction/drug effects
18.
Leukemia ; 34(7): 1805-1815, 2020 07.
Article in English | MEDLINE | ID: mdl-32518419

ABSTRACT

A subgroup of patients with severe COVID-19 suffers from progression to acute respiratory distress syndrome and multiorgan failure. These patients present with progressive hyperinflammation governed by proinflammatory cytokines. An interdisciplinary COVID-19 work flow was established to detect patients with imminent or full blown hyperinflammation. Using a newly developed COVID-19 Inflammation Score (CIS), patients were prospectively stratified for targeted inhibition of cytokine signalling by the Janus Kinase 1/2 inhibitor ruxolitinib (Rux). Patients were treated with efficacy/toxicity guided step up dosing up to 14 days. Retrospective analysis of CIS reduction and clinical outcome was performed. Out of 105 patients treated between March 30th and April 15th, 2020, 14 patients with a CIS ≥ 10 out of 16 points received Rux over a median of 9 days with a median cumulative dose of 135 mg. A total of 12/14 patients achieved significant reduction of CIS by ≥25% on day 7 with sustained clinical improvement in 11/14 patients without short term red flag warnings of Rux-induced toxicity. Rux treatment for COVID-19 in patients with hyperinflammation is shown to be safe with signals of efficacy in this pilot case series for CRS-intervention to prevent or overcome multiorgan failure. A multicenter phase-II clinical trial has been initiated (NCT04338958).


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/drug therapy , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Pneumonia, Viral/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Severe Acute Respiratory Syndrome/drug therapy , Adult , Aged , Aged, 80 and over , Betacoronavirus/drug effects , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Clinical Trials as Topic , Coronavirus Infections/enzymology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokine Release Syndrome/enzymology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/immunology , Drug Administration Schedule , Female , Gene Expression Regulation , Humans , Immunity, Innate/drug effects , Inflammation , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Male , Middle Aged , Nitriles , Pandemics , Patient Safety , Pneumonia, Viral/enzymology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Pyrimidines , Retrospective Studies , SARS-CoV-2 , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/virology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Treatment Outcome
19.
Biochem Pharmacol ; 178: 114103, 2020 08.
Article in English | MEDLINE | ID: mdl-32562787

ABSTRACT

Janus kinase (JAK) inhibitors (also termed Jakinibs) constitute a family of small drugs that target various isoforms of JAKs (JAK1, JAK2, JAK3 and/or tyrosine kinase 2 (Tyk2)). They exert anti-inflammatory properties linked, in part, to the modulation of the activation state of pro-inflammatory M1 macrophages. The exact impact of JAK inhibitors on a wider spectrum of activation states of macrophages is however still to be determined, especially in the context of disorders involving concomitant activation of pro-inflammatory M1 macrophages and profibrotic M2 macrophages. This is especially the case in autoimmune pulmonary fibrosis like scleroderma-associated interstitial lung disease (ILD), in which M1 and M2 macrophages play a key pathogenic role. In this study, we directly compared the anti-inflammatory and anti-fibrotic effects of three JAK inhibitors (ruxolitinib (JAK2/1 inhibitor); tofacitinib (JAK3/2 inhibitor) and itacitinib (JAK1 inhibitor)) on five different activation states of primary human monocyte-derived macrophages (MDM). These three JAK inhibitors exert anti-inflammatory properties towards macrophages, as demonstrated by the down-expression of key polarization markers (CD86, MHCII, TLR4) and the limited secretion of key pro-inflammatory cytokines (CXCL10, IL-6 and TNFα) in M1 macrophages activated by IFNγ and LPS or by IFNγ alone. We also highlighted that these JAK inhibitors can limit M2a activation of macrophages induced by IL-4 and IL-13, as notably demonstrated by the down-regulation of the M2a associated surface marker CD206 and of the secretion of CCL18. Moreover, these JAK inhibitors reduced the expression of markers such as CXCL13, MARCO and SOCS3 in alternatively activated macrophages induced by IL-10 and dexamethasone (M2c + dex) or IL-10 alone (M2c MDM). For all polarization states, Jakinibs with inhibitory properties over JAK2 had the highest effects, at both 1 µM or 0.1 µM. Based on these in vitro results, we also explored the effects of JAK2/1 inhibition by ruxolitinib in vivo, on mouse macrophages in a model of HOCl-induced ILD, that mimics scleroderma-associated ILD. In this model, we showed that ruxolitinib significantly prevented the upregulation of pro-inflammatory M1 markers (TNFα, CXCL10, NOS2) and pro-fibrotic M2 markers (Arg1 and Chi3L3). These results were associated with an improvement of skin and pulmonary involvement. Overall, our results suggest that the combined anti-inflammatory and anti-fibrotic properties of JAK2/1 inhibitors could be relevant to target lung macrophages in autoimmune and inflammatory pulmonary disorders that have no efficient disease modifying drugs to date.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Lung Diseases, Interstitial/drug therapy , Macrophages/drug effects , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Scleroderma, Systemic/drug therapy , Animals , Cell Differentiation , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Female , Gene Expression Regulation , Hypochlorous Acid/administration & dosage , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/genetics , Janus Kinase 3/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung Diseases, Interstitial/chemically induced , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/pathology , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Nitriles , Primary Cell Culture , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/immunology
20.
Expert Rev Hematol ; 13(5): 519-532, 2020 05.
Article in English | MEDLINE | ID: mdl-32249631

ABSTRACT

Introduction: The traditional therapeutic modalities to manage SR-acute GVHD have focused on the inhibition of the alloreactive T-cell response, while in the setting of SR-chronic GVHD the focus has been on a combination of T-cell and B-cell targeting strategies. However, new therapeutic modalities have shown promise. The purpose of this review is to summarize the current treatment landscape of SR-acute and chronic GVHD.Areas covered: A systematic search of MEDLINE, EMBASE, and clinicaltrials.gov databases for published articles, abstracts, and clinical trials pertaining to available therapeutic modalities for SR-acute and SR-chronic GVHD was conducted. Also highlighted is a number of ongoing clinical trials in both SR-acute and SR-chronic GVHD with strategies targeting the JAK-1/2 pathway, the Treg:Tcon ratio, the immunomodulation mediated by mesenchymal stem cells, and the gut microbiome, among others. Expert opinion: Ruxolitinib has emerged as the preferred therapeutic modality for SR-acute GVHD, with alpha-1-antitrypsin and extracorporeal photophoresis (ECP) being reasonable alternatives. Ruxolitinib and Ibrutinib are among the preferred options for SR-chronic GVHD, with ECP being a viable alternative particularly if the skin is involved. A number of novel therapeutic modalities, including those enhancing the activity of regulatory T-cells have shown great promise in early phase trials of SR-chronic GVHD.


Subject(s)
Adenine/analogs & derivatives , Graft vs Host Disease/therapy , Photopheresis , Piperidines/therapeutic use , Pyrazoles/therapeutic use , alpha 1-Antitrypsin/therapeutic use , Acute Disease , Adenine/therapeutic use , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Chronic Disease , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Immunomodulation , Janus Kinase 1/immunology , Janus Kinase 2/immunology , Nitriles , Pyrimidines , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL