1
|
Park DJ, Joo YB, Nam E, Lee J, Bang SY, Lee HS, Bae SC. Exploring potential multiple molecular biomarkers that predict treatment response in patients with lupus nephritis. Sci Rep 2024; 14:31422. [PMID: 39733104 DOI: 10.1038/s41598-024-83057-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Limited knowledge exists regarding biomarkers that predict treatment response in Lupus nephritis (LN). We aimed to identify potential molecular biomarkers to predict treatment response in patients with LN. We enrolled 66 patients with active LN who underwent renal biopsy upon enrollment. Serum and urine samples were collected longitudinally, and we measured 12 biomarkers in each sample using a multiplex immunofluorescence assay. These biomarkers included monocyte chemoattractant protein-1 (MCP-1), interferon gamma-induced protein 10 (IP-10), interferon-γ (IFN-γ), interleukin 6 (IL-6), interleukin 16 (IL-16), interleukin 17 (IL-17), interleukin 23 (IL-23), tumor necrosis factor receptor II (TNF-RII), vascular cell adhesion molecule 1 (VCAM-1), retinol-binding protein 4 (RBP 4), vitamin D binding protein (VDBP), and neutrophil gelatinase-associated lipocalin (NGAL). Patients were categorized into two groups based on their 1-year treatment response to Mycophenolate mofetil (MMF)-based therapy: 50 responders and 16 non-responders. Only urine IL-17 (uIL-17) showed baseline level differences between the two groups, with higher in responders. In ROC curve analyses assessing the predictive performance of biomarkers, baseline uIL-17 and changes in uIL-6 and uIL-23 levels at 3 months could predict the 1-year treatment response, showing AUC values of 0.70 (95% CI 0.54-0.87), 0.70 (0.54-0.86), and 0.71 (0.57-0.85), respectively. Combining uIL-6 and uIL-23 into a model improved predictability, achieving an AUC of 0.75 (0.61-0.90). Baseline uIL-17 levels and early changes in uIL-6 and uIL-23 could serve as potential biomarkers to predict 1-year treatment response in lupus nephritis patients receiving MMF-based therapy.
Collapse
Affiliation(s)
- Dae Jin Park
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea
| | - Young Bin Joo
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea
| | - Eunwoo Nam
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea
| | - Jiyoung Lee
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea
| | - So-Young Bang
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea
| | - Hye-Soon Lee
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea.
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea.
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-Gu, Seoul, 04763, Korea.
- Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, Korea.
| |
Collapse
|
2
|
Klotsman M, Anderson WH, Wyatt D, Lewis T, Theus N, Santoro D. Treatment of moderate-to-severe canine atopic dermatitis with modified-release mycophenolate (OKV-1001): A pilot open-label, single-arm multicentric clinical trial. Vet Dermatol 2024; 35:652-661. [PMID: 39129671 DOI: 10.1111/vde.13283] [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: 03/13/2024] [Revised: 05/26/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Mycophenolate is an immunomodulating agent successfully used for the treatment of moderate-to-severe atopic dermatitis (AD) in people. Mycophenolate is an effective steroid-sparing treatment option for use in dogs with inflammatory skin diseases. OBJECTIVE To evaluate whether once-daily modified-release mycophenolate (OKV-1001) is safe and effective for treating moderate-to-severe canine AD. ANIMALS Client-owned atopic dogs (n = 9) were enrolled. MATERIALS AND METHODS In an open-label multicentre pilot study, OKV-1001 (30 mg/kg every 24 h) was given orally for ≤84 days. Concomitant tapering doses of glucocorticoids were administered up to Day (D)28. Clinicians assessed Canine Atopic Dermatitis Extent and Severity Index, 4th iteration (CADESI-04) on D0, D14, D28, D56 and D84. Body weight and clinical pathological parameters were measured at baseline and at the end of the study. RESULTS Treatment with OKV-1001 combined with glucocorticoids significantly reduced the severity of AD within two weeks in seven of nine (77.8%) dogs. The mean percentage change from baseline in the CADESI-04 score was 29% (p = 0.009) at D14 (n = 9), 39% (p = 0.008) at D28 (n = 9) and 49% (p = 0.03) at D56 (n = 7) at which point glucocorticoids had been withdrawn. In two dogs the improvement in CADESI-04 was 62% and 23% (respectively) on D84. No significant adverse events including clinical pathological findings were reported. CONCLUSIONS AND CLINICAL RELEVANCE Modified-release mycophenolate (OKV-1001) may represent a promising alternative treatment option for dogs with moderate-to-severe AD. The safety and efficacy profile of OKV-1001 will need to be established in larger, placebo-controlled clinical trials.
Collapse
Affiliation(s)
| | - Wayne H Anderson
- Okava Pharmaceuticals, San Francisco, California, USA
- Pulmonary and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Tom Lewis
- Dermatology for Animals, Gilbert, Arizona, USA
| | | | - Domenico Santoro
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
3
|
Kravčenia B, Maślanka T. Mycophenolate Mofetil, an Inhibitor of Inosine Monophosphate Dehydrogenase, and Tofacitinib, a Janus Kinase Inhibitor, Attenuate Airway Inflammation and Hyperresponsiveness in a Mouse Model of Allergic Asthma. Molecules 2024; 29:5293. [PMID: 39598682 PMCID: PMC11597010 DOI: 10.3390/molecules29225293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/19/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Treatment-resistant asthma remains an unresolved clinical problem and a challenge for current medical science. Consequently, there is a growing and urgent need to develop novel or alternative therapeutic options for the treatment of asthma. The research problem raised in this study was to assess and compare mycophenolate mofetil (MMF), an inhibitor of inosine monophosphate dehydrogenase, and tofacitinib (TFB), a Janus kinase inhibitor, for anti-asthmatic properties, and consequently to determine whether these agents may have potential as alternative options for treatment of allergic asthma. For this purpose, we assessed the effect of administration of MMF and TFB on the development of a mouse model of allergic airway inflammation (AAI) and accompanying CD4+ (cluster of differentiation 4) T-cell immune response in the lung-draining mediastinal lymph nodes (MLNs) and lungs, i.e., in the inductive and effector sites, respectively, of the immune response underlying the development of allergic asthma. The results from a histopathological scoring system demonstrated that the administration of MMF and TFB did not prevent or abolish ovalbumin-induced AAI, but strongly attenuated its severity. The pulmonary function tests revealed that the treatment with MMF and TFB significantly reduced methacholine-induced bronchoconstriction. These results indicate that the treatment with TFB and MMF attenuated the development of ovalbumin-induced AAI. The magnitude of the anti-asthmatic effect was comparable between both agents. The study revealed that the impairment of the clonal expansion of effector CD4+ T cells in the MLNs is a critical event in the mechanism underlying the anti-asthmatic effect of MMF and TFB. Apart from this, the findings of the study strongly suggest that the suppression of the interleukin-33/suppression of tumorigenicity-2 signaling pathway may constitute an additional mechanism responsible for producing this effect. In turn, the results indicate that the anti-asthmatic action induced by the studied agents is not mediated by the generation of forkhead box protein 3-expressing CD4+ regulatory T cells. Clinical implication of the results: the results suggest that MMF and TFB may exert anti-asthmatic action, and thus they may be considered therapeutic options for the treatment of allergic asthma cases resistant to conventional/existing treatment.
Collapse
Affiliation(s)
| | - Tomasz Maślanka
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Street 13, 10-719 Olsztyn, Poland;
| |
Collapse
|
4
|
Zhao M, Cheng Y, Gao J, Zhou F. Single-cell mass cytometry in immunological skin diseases. Front Immunol 2024; 15:1401102. [PMID: 39081313 PMCID: PMC11286489 DOI: 10.3389/fimmu.2024.1401102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Immune-related skin diseases represent a collective of dermatological disorders intricately linked to dysfunctional immune system processes. These conditions are primarily characterized by an immoderate activation of the immune system or deviant immune responses, involving diverse immune components including immune cells, antibodies, and inflammatory mediators. However, the precise molecular dysregulation underlying numerous individual cases of these diseases and unique subsets respond under disease conditions remains elusive. Comprehending the mechanisms and determinants governing the homeostasis and functionality of diseases could offer potential therapeutic opportunities for intervention. Mass cytometry enables precise and high-throughput quantitative measurement of proteins within individual cells by utilizing antibodies labeled with rare heavy metal isotopes. Imaging mass cytometry employs mass spectrometry to obtain spatial information on cell-to-cell interactions within tissue sections, simultaneously utilizing more than 40 markers. The application of single-cell mass cytometry presents a unique opportunity to conduct highly multiplexed analysis at the single-cell level, thereby revolutionizing our understanding of cell population heterogeneity and hierarchy, cellular states, multiplexed signaling pathways, proteolysis products, and mRNA transcripts specifically in the context of many autoimmune diseases. This information holds the potential to offer novel approaches for the diagnosis, prognostic assessment, and monitoring responses to treatment, thereby enriching our strategies in managing the respective conditions. This review summarizes the present-day utilization of single-cell mass cytometry in studying immune-related skin diseases, highlighting its advantages and limitations. This technique will become increasingly prevalent in conducting extensive investigations into these disorders, ultimately yielding significant contributions to their accurate diagnosis and efficacious therapeutic interventions.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yuqi Cheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Jinping Gao
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Fusheng Zhou
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| |
Collapse
|
5
|
Lin L, Ren R, Xiong Q, Zheng C, Yang B, Wang H. Remodeling of T-cell mitochondrial metabolism to treat autoimmune diseases. Autoimmun Rev 2024; 23:103583. [PMID: 39084278 DOI: 10.1016/j.autrev.2024.103583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
T cells are key drivers of the pathogenesis of autoimmune diseases by producing cytokines, stimulating the generation of autoantibodies, and mediating tissue and cell damage. Distinct mitochondrial metabolic pathways govern the direction of T-cell differentiation and function and rely on specific nutrients and metabolic enzymes. Metabolic substrate uptake and mitochondrial metabolism form the foundational elements for T-cell activation, proliferation, differentiation, and effector function, contributing to the dynamic interplay between immunological signals and mitochondrial metabolism in coordinating adaptive immunity. Perturbations in substrate availability and enzyme activity may impair T-cell immunosuppressive function, fostering autoreactive responses and disrupting immune homeostasis, ultimately contributing to autoimmune disease pathogenesis. A growing body of studies has explored how metabolic processes regulate the function of diverse T-cell subsets in autoimmune diseases such as systemic lupus erythematosus (SLE), multiple sclerosis (MS), autoimmune hepatitis (AIH), inflammatory bowel disease (IBD), and psoriasis. This review describes the coordination of T-cell biology by mitochondrial metabolism, including the electron transport chain (ETC), oxidative phosphorylation, amino acid metabolism, fatty acid metabolism, and one‑carbon metabolism. This study elucidated the intricate crosstalk between mitochondrial metabolic programs, signal transduction pathways, and transcription factors. This review summarizes potential therapeutic targets for T-cell mitochondrial metabolism and signaling in autoimmune diseases, providing insights for future studies.
Collapse
Affiliation(s)
- Liyan Lin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruyu Ren
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiao Xiong
- Department of Infectious Disease, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Bin Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Wang Y, Huang T, Gu J, Lu L. Targeting the metabolism of tumor-infiltrating regulatory T cells. Trends Immunol 2023:S1471-4906(23)00109-6. [PMID: 37442660 DOI: 10.1016/j.it.2023.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023]
Abstract
Although targeting the tumor metabolism is performed in cooperation with immunotherapy in the era of precision oncology, ignorance of immune cells' metabolism has resulted in unstable antitumor responses. Tumor-infiltrating regulatory T cells (TI-Tregs) are unique, overcoming the hypoxic, acidic, and nutrient-deficient tumor microenvironments (TMEs) and maintaining immunosuppressive functions. However, secondary autoimmunity caused by systemic Treg depletion remains the 'Sword of Damocles' for current Treg-targeted therapies. In this opinion piece, we propose that metabolically reprogrammed TI-Tregs might represent an obstacle to cancer therapies. Indeed, metabolism-based Treg-targeted therapy might provide higher selectivity for clearing TI-Tregs than traditional kinase/checkpoint inhibitors and chemokine/chemokine receptor blockade; it might also restore the efficacy of targeting the tumor metabolism and eliminate certain metabolic barriers to immunotherapy.
Collapse
Affiliation(s)
- Yiming Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Tianning Huang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Jian Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Ling Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
7
|
Yang J, Li S. Molecular mechanism of Hedyotis Diffusae Herba in the treatment of lupus nephritis based on network pharmacology. Front Pharmacol 2023; 14:1118804. [PMID: 37361229 PMCID: PMC10285311 DOI: 10.3389/fphar.2023.1118804] [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: 12/08/2022] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Aims: To determine the bioactive components of Hedyotis Diffusae Herba (HDH) and the targets in treating lupus nephritis (LN), and so as to elucidate the protective mechanism of HDH against LN. Methods and results: An aggregate of 147 drug targets and 162 LN targets were obtained from online databases, with 23 overlapped targets being determined as potential therapeutic targets of HDH against LN. Through centrality analysis, TNF, VEGFA and JUN were screened as core targets. And the bindings of TNF with stigmasterol, TNF with quercetin, and VEGFA with quercetin were further validated by molecular docking. By conducting Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses for drug targets, disease targets and the shared targets, TNF signaling pathway, Toll-like receptor signaling pathway, NF-kappa B signaling pathway and HIF-1 signaling pathway, etc., were found in all these three lists, indicating the potential mechanism of HDH in the treatment of LN. Conclusion: HDH may ameliorate the renal injury in LN by targeting multi-targets and multi-pathways, including TNF signaling pathway, NF-kappa B signaling pathway, HIF-1 signaling pathway and so on, which provided novel insights into further researches of the drug discovery in LN.
Collapse
Affiliation(s)
- Jinfei Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Siying Li
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
8
|
Petrić M, Radić M. Is Th17-Targeted Therapy Effective in Systemic Lupus Erythematosus? Curr Issues Mol Biol 2023; 45:4331-4343. [PMID: 37232744 DOI: 10.3390/cimb45050275] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a broad spectrum of clinical manifestations. The proposed pathophysiological hypotheses of SLE are numerous, involving both innate and adaptive abnormal immune responses. SLE is characterized by the overproduction of different autoantibodies that form immune complexes, which cause damage in different organs. Current therapeutic modalities are anti-inflammatory and immunosuppressive. In the last decade, we have witnessed the development of many biologicals targeting different cytokines and other molecules. One of them is interleukin-17 (IL-17), a central cytokine of a proinflammatory process that is mediated by a group of helper T cells called Th17. Direct inhibitors of IL-17 are used in psoriatic arthritis, spondyloarthritis, and other diseases. Evidence about the therapeutic potential of Th17-targeted therapies in SLE is scarce, and probably the most promising is related to lupus nephritis. As SLE is a complex heterogeneous disease with different cytokines involved in its pathogenesis, it is highly unlikely that inhibition of only one molecule, such as IL-17, will be effective in the treatment of all clinical manifestations. Future studies should identify SLE patients that are eligible for Th17-targeted therapy.
Collapse
Affiliation(s)
- Marin Petrić
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, University Hospital of Split, Center of Excellence for Systemic Sclerosis Ministry of Health Republic of Croatia, Šoltanska 1, 21000 Split, Croatia
| | - Mislav Radić
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, University Hospital of Split, Center of Excellence for Systemic Sclerosis Ministry of Health Republic of Croatia, Šoltanska 1, 21000 Split, Croatia
- Department of Internal Medicine, School of Medicine, University of Split, Šoltanska 2, 21000 Split, Croatia
| |
Collapse
|
9
|
Gibbs LC, Oviedo JM, Ondigo BN, Fairfax KC. Maternal infection causes dysfunctional BCR signaling in male offspring due to aberrant Xist expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528357. [PMID: 36824836 PMCID: PMC9948949 DOI: 10.1101/2023.02.13.528357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Infections during pregnancy with pathogens such as helminths correlate with altered immune responses to common childhood immunizations. However, the molecular mechanisms that underlie this remain unknown. Using our murine model of maternal schistosomiasis, when immunized, males from infected mothers had a lower frequency of antigen-specific germinal center B cells and downregulation of transcripts downstream of BCR signaling compared to males from uninfected mothers. This is driven by a reduction in developing B cell populations within the bone marrow of pups from infected mothers. Males from infected mothers were impacted to a greater extent than their female littermate counterparts. We found this defect to be caused by aberrant expression of the long non-coding RNA Xist in males leading to dysregulated Igα expression on developing B cells. This, for the first time, links dysfunctional BCR signaling with Xist expression, while also proposing a detrimental function for Xist expression in males.
Collapse
Affiliation(s)
- Lisa C. Gibbs
- Department of Pathology, University of Utah; Salt Lake City, UT, United States
| | - Juan M. Oviedo
- Department of Pathology, University of Utah; Salt Lake City, UT, United States
| | | | - Keke C. Fairfax
- Department of Pathology, University of Utah; Salt Lake City, UT, United States
| |
Collapse
|
10
|
Matsumoto K, Suzuki K, Yoshida H, Magi M, Matsumoto Y, Noguchi-Sasaki M, Yoshimoto K, Takeuchi T, Kaneko Y. Distinct gene signatures of monocytes and B cells in patients with giant cell arteritis: a longitudinal transcriptome analysis. Arthritis Res Ther 2023; 25:1. [PMID: 36597161 PMCID: PMC9809009 DOI: 10.1186/s13075-022-02982-9] [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: 09/11/2022] [Accepted: 12/20/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Giant cell arteritis (GCA) is a primary large-vessel vasculitis (LVV) of unknown origin. Its management is a challenge due to the late onset of disease symptoms and frequent relapse; therefore, clarifying the pathophysiology of GCA is essential to improving treatment. This study aimed to identify the transition of molecular signatures in immune cells relevant to GCA pathogenesis by analyzing longitudinal transcriptome data in patients. METHODS We analyzed the whole blood transcriptome of treatment-naive patients with GCA, patients with Takayasu arteritis (TAK), age-matched, old healthy controls (HCs), and young HCs. Characteristic genes for GCA were identified, and the longitudinal transition of those genes was analyzed using cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT). RESULTS Repeated measures analysis of variance revealed 739 differentially expressed genes among all patients and HCs. Of the 739 genes, 15 were characteristically upregulated and 36 were downregulated in patients with GCA compared to those with TAK and HCs. Pathway enrichment analysis showed that downregulated genes in GCA were associated with B cell activation. CIBERSORT analysis revealed that upregulation of "M0-macrophages" and downregulation of B cells were characteristic of GCA. Upregulation of "M0-macrophages" reflects the activation of monocytes in GCA toward M0-like phenotypes, which persisted under 6 weeks of treatment. Combined treatment with prednisolone and an interleukin-6 receptor antagonist normalized molecular profiles more efficiently than prednisolone monotherapy. CONCLUSIONS Gene signatures of monocyte activation and B cell inactivation were characteristic of GCA and associated with treatment response.
Collapse
Affiliation(s)
- Kotaro Matsumoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Tokyo, Shinjuku-ku, Japan.
| | - Katsuya Suzuki
- grid.26091.3c0000 0004 1936 9959Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Tokyo, Shinjuku-ku Japan
| | - Hiroto Yoshida
- grid.515733.60000 0004 1756 470XChugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa Japan
| | - Mayu Magi
- grid.515733.60000 0004 1756 470XChugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa Japan
| | - Yoshihiro Matsumoto
- grid.515733.60000 0004 1756 470XChugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa Japan
| | - Mariko Noguchi-Sasaki
- grid.515733.60000 0004 1756 470XChugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa Japan
| | - Keiko Yoshimoto
- grid.26091.3c0000 0004 1936 9959Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Tokyo, Shinjuku-ku Japan
| | - Tsutomu Takeuchi
- grid.26091.3c0000 0004 1936 9959Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Tokyo, Shinjuku-ku Japan
| | - Yuko Kaneko
- grid.26091.3c0000 0004 1936 9959Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Tokyo, Shinjuku-ku Japan
| |
Collapse
|
11
|
Zhou X, Wang Y, Huang B, Feng R, Zhou X, Li C, Zhang X, Shao M, Gan Y, Jin Y, An Y, Xiao X, Wang S, Liu Q, Cheng G, Zhu F, Zhang K, Wang N, Xing X, Li R, Li Y, Liu Y, Lu D, Sun X, Li Z, Liu Y, He J. Dynamics of T follicular helper cells in patients with rheumatic diseases and subsequent antibody responses in a three-dose immunization regimen of CoronaVac. Immunology 2023; 168:184-197. [PMID: 36057099 DOI: 10.1111/imm.13572] [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: 06/08/2022] [Accepted: 08/28/2022] [Indexed: 12/30/2022] Open
Abstract
Given increased acceptance of the CoronaVac, there is an unmet need to assess the safety and immunogenic changes of CoronaVac in patients with rheumatic diseases (RD). Here we comprehensively analysed humoral and cellular responses in patient with RD after a three-dose immunization regimen of CoronaVac. RD patients with stable condition and/or low disease activity (n = 40) or healthy controls (n = 40) were assigned in a 1:1 ratio to receive CoronaVac (Sinovac). The prevalence of anti-receptor binding domain (RBD) antibodies and neutralizing antibodies was similar between healthy control (HC) and RD patients after the second and the third vaccination. However, the titers of anti-RBD IgG and neutralizing antibodies were significantly lower in RD patients compared to HCs (p < 0.05), which was associated with an impaired T follicular helper (Tfh) cell response. Among RD patients, those who generated an antibody response displayed a significantly higher Tfh cells compared to those who failed after the first and the second vaccination (p < 0.05). Interestingly, subjects with a negative serological response displayed a similar Tfh memory response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-derived peptides as their anti-RBD IgG positive counterpart, and all (4/4) of the non-responders in HCs, and 62.5% (5/8) of the non-responders in patients with RD displayed a positive serological response following the third dose. No serious adverse events were observed. In conclusion, our findings support SARS-CoV-2 vaccination in patients with RD with stable and/or low disease activity. The impaired ability in generating vaccine-specific antibodies in patients with RD was associated with a reduction in Tfh cells induction. The window of vaccination times still needs to be explored in future studies. Clinical trial registration: This trial was registered with ChiCTR2100049138.
Collapse
Affiliation(s)
- Xingyu Zhou
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yifan Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Bo Huang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Ruiling Feng
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xinyao Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chun Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xia Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Miao Shao
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuzhou Gan
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuebo Jin
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuan An
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xian Xiao
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Shiyang Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Qinghong Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Gong Cheng
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Fengyunzhi Zhu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Kai Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Naidi Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xiaoyan Xing
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Ru Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuhui Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yisi Liu
- The First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Dan Lu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yudong Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
12
|
Xu L, Li Y, Ji J, Lai Y, Chen J, Ding T, Li H, Ding B, Ge W. The anti-inflammatory effects of Hedyotis diffusa Willd on SLE with STAT3 as a key target. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115597. [PMID: 35940466 DOI: 10.1016/j.jep.2022.115597] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedyotis diffusa Willd, also named Scleromitrion diffusum (Willd.) R.J. Wang, is one medical herb, which has been traditionally used by the She nationality in China. And H. diffusa represents a beneficial effect on Systemic lupus erythematosus (SLE) treatment in clinic. AIM OF THE STUDY The underlying mechanisms of the protective effects of H. diffusa on SLE remain unclear. In this study, we treated MRL/lpr mice with H. diffusa water extract (HDW) to assess its therapeutic effects and verified its regulating signalling pathway through cytological experiments. MATERIALS AND METHODS In the present study, the constituents of HDW were analysed through ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) and SCIEX OS software. The protective activity and underlying mechanisms were studied in a MRL/lpr lupus mouse model. The blood cells, autoantibodies, metabolites and the cytokines in serum were identified with a hematology analyzer, specific ELISA kit, GC/MS system and cytometric assays. The histological and immunohistochemical analysis were engaged in the morphologic, and the expression and translocation of the crucial protein observation. The dual luciferase reporter assay was applied to identifying the regulative activity of HDW. The transcription and translation expression of the protein was studied by real-time PCR and Western blot assays. The network pharmacology analysis was employed to predict the IL-6/STAT3 pathway regulators and the screen the STAT3 inhibitors in HDW. RESULTS The results revealed the capability of HDW to attenuate the production of autoantibodies, secretion of inflammatory cytokines (IL-6 and IFN-γ), and suppressed the IgG and C3 deposition, the development of glomerular lesions in MRL/lpr mice. Serum metabolomics study showed the improvement in serum metabolites, especially aminoacyl-tRNA biosynthesis, by HDW. IL-6 was clarified to be highly associated with the significantly changed metabolites in network analysis. We further demonstrated the effects of HDW on the IL-6/STAT3 pathway in vivo and in vitro. CONCLUSIONS This study suggested that HDW exerts a therapeutic effect in SLE model mice by suppressing the IL-6/STAT3 pathway.
Collapse
Affiliation(s)
- Li Xu
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Ying Li
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Jinjun Ji
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Yahui Lai
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Jing Chen
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Tao Ding
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Haichang Li
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Bin Ding
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Weihong Ge
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| |
Collapse
|
13
|
Kim K, Narasimhan M, Mahimainathan L, Zhang R, Araj E, Kim E, Tharpe W, Greenberg BM, Greenberg DE, Li QZ, Cheng CA, Sarode R, Malladi S, Muthukumar A. Translation suppression underlies the restrained COVID-19 mRNA vaccine response in the high-risk immunocompromised group. Front Immunol 2022; 13:1020165. [DOI: 10.3389/fimmu.2022.1020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundImmunocompromised (IC) patients show diminished immune response to COVID-19 mRNA vaccines (Co-mV). To date, there is no ‘empirical’ evidence to link the perturbation of translation, a rate-limiting step for mRNA vaccine efficiency (VE), to the dampened response of Co-mV.Materials and methodsImpact of immunosuppressants (ISs), tacrolimus (T), mycophenolate (M), rapamycin/sirolimus (S), and their combinations on Pfizer Co-mV translation were determined by the Spike (Sp) protein expression following Co-mV transfection in HEK293 cells. In vivo impact of ISs on SARS-CoV-2 spike specific antigen (SpAg) and associated antibody levels (IgGSp) in serum were assessed in Balb/c mice after two doses (2D) of the Pfizer vaccine. Spike Ag and IgGSp levels were assessed in 259 IC patients and 50 healthy controls (HC) who received 2D of Pfizer or Moderna Co-mV as well as in 67 immunosuppressed solid organ transplant (SOT) patients and 843 non-transplanted (NT) subjects following three doses (3D) of Co-mV. Higher Co-mV concentrations and transient drug holidays were evaluated.ResultsWe observed significantly lower IgGSP response in IC patients (p<0.0001) compared to their matched controls in 2D and 3D Co-mV groups. IC patients on M or S showed a profound dampening of IgGSP response relative to those that were not on these drugs. M and S, when used individually or in combination, significantly attenuated the Co-mV-induced Sp expression, whereas T did not exert significant influence. Sirolimus combo pretreatment in vivo significantly attenuated the Co-mV induced IgMSp and IgGSp production, which correlated with a decreasing trend in the early levels (after day 1) of Co-mV induced Sp immunogen levels. Neither higher Co-mV concentrations (6μg) nor withholding S for 1-day could overcome the inhibition of Sp protein levels. Interestingly, 3-days S holiday or using T alone rescued Sp levels in vitro.ConclusionsThis is the first study to demonstrate that ISs, sirolimus and mycophenolate inhibited Co-mV-induced Sp protein synthesis via translation repression. Selective use of tacrolimus or drug holiday of sirolimus can be a potential means to rescue translation-dependent Sp protein production. These findings lay a strong foundation for guiding future studies aimed at improving Co-mV responses in high-risk IC patients.
Collapse
|
14
|
Persia FA, Abba R, Pascual LI, Hapon MB, Mackern-Oberti JP, Gamarra-Luques C. Prosopis strombulifera aqueous extract reduces T cell response and ameliorates type I diabetes in NOD mice. J Tradit Complement Med 2022; 13:20-29. [PMID: 36685075 PMCID: PMC9845655 DOI: 10.1016/j.jtcme.2022.10.001] [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: 02/23/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022] Open
Abstract
Background New products with tolerogenic properties on T cell response are necessary to improve current efficacy, cost and side effects of immunosuppressants. Prosopis strombulifera aqueous extract (PsAE) have reported cytotoxic, antitumoral, antiatherogenic and antileishmanial activities, containing phytochemicals with immune-related activities. Despite these, there are no previous studies with respect to PsAE suppressive properties over T cell proliferation and their function. Goal Because of previous antecedents, this study aims to evaluate the effect of PsAE on T cell activation, proliferation, cytokine production, and to investigate its effect over an in vivo model of type 1 diabetes (T1D). Experimental procedure Splenocytes and sorted CD4+/CD8+ from wild type C57BL/6 mice were cultured to determine activation, IFN-γ release and T-cell proliferation after polyclonal stimulation. NOD (non-obese diabetic) mice were used to study the effects of orally administered extract on glycemia, insulitis stages and perforin-1 (PRF-1)/granzyme-B (GRZ-B) expression. Results In primary cultures, the plant extract impairs T cell activation, decreases IFN-γ release, and reduces proliferation after different polyclonal stimuli. In vivo, PsAE improves NOD mice glycemic levels and T1D progression by diminution of pancreas insulitis and reduction of PRF-1 and GRZ-B mRNA expression. To our knowledge, this is the first report characterizing the therapeutic properties of PsAE on T cell activation. Conclusion The current work provides evidence about in vitro and in vivo immunosuppressive effects of PsAE and promotes this plant extract as a complementary and alternative treatment in autoimmune T-cell mediated diseases as T1D.
Collapse
Affiliation(s)
- Fabio Andrés Persia
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo, CCT Mendoza CONICET, Argentina,Facultad de Ciencias Médicas, Universidad de Mendoza, Argentina
| | - Romina Abba
- Instituto de Histología y Embriología de Mendoza, CCT Mendoza CONICET, Argentina
| | - Lourdes Inés Pascual
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo, CCT Mendoza CONICET, Argentina
| | - María Belén Hapon
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo, CCT Mendoza CONICET, Argentina,Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Juan Pablo Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo, CCT Mendoza CONICET, Argentina,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Carlos Gamarra-Luques
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo, CCT Mendoza CONICET, Argentina,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina,Corresponding author. Instituto de Medicina y Biología Experimental de Cuyo, Universidad Nacional de Cuyo, CCT Mendoza CONICET, Av. Ruiz Leal s/n. Casilla de Correo 0855, CP5500, Mendoza, Provincia de Mendoza, Argentina.
| |
Collapse
|
15
|
Exploring the Molecular Mechanism of Zhi Bai Di Huang Wan in the Treatment of Systemic Lupus Erythematosus Based on Network Pharmacology and Molecular Docking Techniques. Processes (Basel) 2022. [DOI: 10.3390/pr10101914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: To investigate the molecular mechanism and simulated validation of Zhi Bai Di Huang Pill (ZBDHP) for the treatment of systemic lupus erythematosus (SLE) using network pharmacology and molecular docking techniques. Methods: The active ingredients of ZBDHP were obtained through the TCMSP database and the Canonical SMILES of the active ingredients were queried through Pubchem. The targets of the active ingredients were predicted in the SwissTarget database based on the SMILES. The SLE-related disease targets were obtained through the GeneCards, OMIM and DisGenets databases, and the intersection targets of ZBDHP and SLE were obtained using the Venny 2.1.0 online platform. Intersection targets build a visual protein interaction network (PPI) through the STRING database, and the core targets were identified by network topology analysis. GO analysis and KEGG pathway enrichment analysis of the intersecting targets were performed using the DAVID database. Finally, the molecular docking of the first four active ingredients and the first four core target genes were verified by Pubchem, the PDB database and CB-Dock online molecular docking technology. Results: ZBDHP screened 91 potential active ingredients and 816 potential targets. Among them, 141 genes were intersected by ZBDHP and SLE. The network topology analysis showed that the main active ingredients were Hydroxygenkwanin, Alisol B, asperglaucide, Cerevisterol, etc., and the key target genes were TNF, AKT1, EGFR, STAT3, etc. GO and KEGG enrichment analysis showed that common targets interfere with biological processes or molecular functions such as signal transduction protein phosphorylation, inflammatory response, transmembrane receptor protein tyrosine kinase activity, etc., through multiple signaling pathways, such as pathways in cancer, Kaposi sarcoma-associated herpesvirus infection, the PI3K-Akt signaling pathway, lipid and atherosclerosis, hepatitis B, etc. Molecular docking results showed that the active components of ZBDHP have good binding activity to the core targets of SLE. Conclusions: This study reveals that the ZBDHP treatment of SLE is a complex mechanistic process with multi-components, multi-targets and multi-pathways, and it may play a therapeutic role in SLE by inhibiting the production, proliferation and apoptosis of inflammatory factors. In conclusion, the present study provides a theoretical basis for further research on ZBDHP.
Collapse
|
16
|
Sosa-Hernández VA, Romero-Ramírez S, Cervantes-Díaz R, Carrillo-Vázquez DA, Navarro-Hernandez IC, Whittall-García LP, Absalón-Aguilar A, Vargas-Castro AS, Reyes-Huerta RF, Juárez-Vega G, Meza-Sánchez DE, Ortiz-Navarrete V, Torres-Ruiz J, Mejía-Domínguez NR, Gómez-Martín D, Maravillas-Montero JL. CD11c + T-bet + CD21 hi B Cells Are Negatively Associated With Renal Impairment in Systemic Lupus Erythematosus and Act as a Marker for Nephritis Remission. Front Immunol 2022; 13:892241. [PMID: 35663936 PMCID: PMC9160198 DOI: 10.3389/fimmu.2022.892241] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
Lupus nephritis (LN) is one of the most common manifestations of systemic lupus erythematosus (SLE), characterized by abnormal B cell activation and differentiation to memory or plasma effector cells. However, the role of these cells in the pathogenesis of LN is not fully understood, as well as the effect of induction therapy on B cell subsets, possibly associated with this manifestation, like aged-associated B cells (ABCs). Consequently, we analyzed the molecules defining the ABCs subpopulation (CD11c, T-bet, and CD21) through flow cytometry of blood samples from patients with lupus presenting or not LN, following up a small sub-cohort after six months of induction therapy. The frequency of ABCs resulted higher in LN patients compared to healthy subjects. Unexpectedly, we identified a robust reduction of a CD21hi subset that was almost specific to LN patients. Moreover, several clinical and laboratory lupus features showed strong and significant correlations with this undefined B cell subpopulation. Finally, it was observed that the induction therapy affected not only the frequencies of ABCs and CD21hi subsets but also the phenotype of the CD21hi subset that expressed a higher density of CXCR5. Collectively, our results suggest that ABCs, and more importantly the CD21hi subset, may work to assess therapeutic response since the reduced frequency of CD21hi cells could be associated with the onset of LN.
Collapse
Affiliation(s)
- Víctor A Sosa-Hernández
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico.,Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Sandra Romero-Ramírez
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rodrigo Cervantes-Díaz
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Daniel A Carrillo-Vázquez
- Departamento de Medicina Interna, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Itze C Navarro-Hernandez
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico.,Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Laura P Whittall-García
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Abdiel Absalón-Aguilar
- Departamento de Medicina Interna, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Ana S Vargas-Castro
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Raúl F Reyes-Huerta
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - David E Meza-Sánchez
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Jiram Torres-Ruiz
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diana Gómez-Martín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José L Maravillas-Montero
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
17
|
Assassi S, Volkmann ER, Zheng WJ, Wang X, Wilhalme H, Lyons MA, Roth MD, Tashkin DP. Peripheral blood gene expression profiling shows predictive significance for response to mycophenolate in systemic sclerosis-related interstitial lung disease. Ann Rheum Dis 2022; 81:854-860. [PMID: 35190386 PMCID: PMC9117450 DOI: 10.1136/annrheumdis-2021-221313] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To characterise the peripheral blood cell (PBC) gene expression changes ensuing from mycophenolate mofetil (MMF) or cyclophosphamide (CYC) treatment and to determine the predictive significance of baseline PBC transcript scores for response to immunosuppression in systemic sclerosis (SSc)-related interstitial lung disease (ILD). METHODS PBC RNA samples from baseline and 12-month visits, corresponding to the active treatment period of both arms in Scleroderma Lung Study II, were investigated by global RNA sequencing. Joint models were created to examine the predictive significance of baseline composite modular scores for the course of forced vital capacity (FVC) per cent predicted measurements from 3 to 12 months. RESULTS 134 patients with SSc-ILD (CYC=69 and MMF=65) were investigated. CYC led to an upregulation of erythropoiesis, inflammation and myeloid lineage-related modules and a downregulation of lymphoid lineage-related modules. The modular changes resulting from MMF treatment were more modest and included a downregulation of plasmablast module. In the longitudinal analysis, none of the baseline transcript module scores showed predictive significance for FVC% course in the CYC arm. In contrast, in the MMF arm, higher baseline lymphoid lineage modules predicted better subsequent FVC% course, while higher baseline myeloid lineage and inflammation modules predicted worse subsequent FVC% course. CONCLUSION Consistent with the primary mechanism of action of MMF on lymphocytes, patients with SSc-ILD with higher baseline lymphoid module scores had better FVC% course, while those with higher myeloid cell lineage activation score had poorer FVC% course on MMF.
Collapse
Affiliation(s)
- Shervin Assassi
- Rheumatology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xuan Wang
- Baylor Institute for Immunology Research, Dallas, Texas, USA
| | - Holly Wilhalme
- Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Marka A Lyons
- Rheumatology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael D Roth
- Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Donald P Tashkin
- Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
18
|
Du LJ, Feng YX, He ZX, Huang L, Wang Q, Wen CP, Zhang Y. Norcantharidin ameliorates the development of murine lupus via inhibiting the generation of IL-17 producing cells. Acta Pharmacol Sin 2022; 43:1521-1533. [PMID: 34552214 PMCID: PMC9159996 DOI: 10.1038/s41401-021-00773-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a devastating autoimmune disorder associated with severe organ damage. The abnormality of T cell apoptosis is considered as an important pathogenetic mechanism of SLE. Norcantharidin (NCTD), a derivative of Cantharidin, is an efficacious anti-cancer drug by inhibiting cell proliferation and inducing cell apoptosis. Besides, NCTD has also been proved to protect the function of kidneys, while damaged renal function is the most important predictor of morbidity and mortality in SLE. All these suggest the potential effects of NCTD in SLE treatment. In this study we investigated whether NCTD exerted therapeutic effects in a mouse SLE model. Lupus prone female MRL/lpr mice were treated with NCTD (1, 2 mg·kg-1·d-1, ip) for 8 weeks. We showed that NCTD administration significantly decreased mortality rate, diminished the expression of anti-dsDNA IgG antibody, a diagnostic marker for SLE, as well as restored renal structure and function in MRL/lpr mice. Moreover, NCTD administration dose-dependently inhibited lymphoproliferation and T cell accumulation in the spleens of MRL/lpr mice. We further revealed that NCTD specifically inhibited DN T cell proliferation and Th17 cell differentiation both via blocking activation of signal transducer and activator of transcription 3 (STAT3) signaling pathway. On the other hand, NCTD did not affect T cell apoptosis in MRL/lpr mice. Taken together, our data suggest that NCTD may be as a promising therapeutic drug through targeting T cells for the treatment of SLE.
Collapse
Affiliation(s)
- Li-jun Du
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Yu-xiang Feng
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Zhi-xing He
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Lin Huang
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Qiao Wang
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Cheng-ping Wen
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Yun Zhang
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| |
Collapse
|
19
|
Mehta RS, Saliba RM, Hayase E, Jenq RR, Abraham S, Rashid A, Rondon G, Al-Atrash G, Bashir Q, Hosing CM, Kebriaei P, Khouri I, Marin D, Nieto Y, Olson A, Oran B, Popat UR, Qazilbash MH, Ramdial J, Srour S, Champlin RE, Rezvani K, Shpall EJ, Alousi AM. Mycophenolate Mofetil: A Friend or a Foe with PTCy and Tacrolimus Prophylaxis in HLA-Matched donors? Transplant Cell Ther 2022; 28:500.e1-500.e10. [PMID: 35662592 DOI: 10.1016/j.jtct.2022.05.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/02/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
Abstract
Adapted from the haploidentical literature, post-transplantation cyclophosphamide (PTCy) is increasingly being used with HLA-matched donors, generally with a calcineurin inhibitor, such as tacrolimus (Tac) with or without mycophenolate mofetil (MMF). Owing to its immunosuppressive, potentially antitumor, and antimicrobial properties, MMF is an attractive drug; however, it remains unclear how much benefit is gained when used with PTCy/Tac. To assess that, we compared PTCy/Tac (n=242) to PTCy/Tac/MMF (n= 144) in recipients of HLA-matched donors. In multivariate analysis, the PTCy/Tac/MMF group had a significantly higher risk of grade II-IV acute graft-versus-host disease (GVHD; hazard ratio (HR) 2.1, 95% confidence interval (CI) 1.6-2.8, p<0.001), and steroid-refractory/dependent acute GVHD (HR 4.8, 95% CI 2.4-9.6, p<0.001), yet a significantly lower risk of relapse (HR 0.5, 95% CI, 0.3-0.9, p=0.009) and better progression-free survival (PFS; HR 0.7, 95% CI 0.5-0.9, p=0.04). There was no difference in the risk of grade III-IV acute GVHD, chronic GVHD, non-relapse mortality, or overall survival. MMF was associated with prolonged neutrophil engraftment by 2 days, and a higher risk of bacterial infections. In an exploratory stool microbiome analysis (n=16), we noted a higher relative abundance of β-glucuronidase-producing bacteria in the MMF group, which may have a role in the pathogenesis of MMF-related GVHD. Our data suggest that the addition of MMF to PTCy/Tac for HLA-matched donor HCT does not provide any advantage for GVHD prevention. Further studies are needed to decipher this mechanism, and understand its role with PTCy-based prophylaxis.
Collapse
Affiliation(s)
- Rohtesh S Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Rima M Saliba
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Eiko Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert R Jenq
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Susan Abraham
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Asif Rashid
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Chitra M Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Issa Khouri
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amanda Olson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Betul Oran
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Uday R Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Muzaffar H Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amin M Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
20
|
Schrezenmeier E, Rincon-Arevalo H, Jens A, Stefanski AL, Hammett C, Osmanodja B, Koch N, Zukunft B, Beck J, Oellerich M, Proß V, Stahl C, Choi M, Bachmann F, Liefeldt L, Glander P, Schütz E, Bornemann-Kolatzki K, López del Moral C, Schrezenmeier H, Ludwig C, Jahrsdörfer B, Eckardt KU, Lachmann N, Kotsch K, Dörner T, Halleck F, Sattler A, Budde K. Temporary antimetabolite treatment hold boosts SARS-CoV-2 vaccination-specific humoral and cellular immunity in kidney transplant recipients. JCI Insight 2022; 7:e157836. [PMID: 35349490 PMCID: PMC9090237 DOI: 10.1172/jci.insight.157836] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/24/2022] [Indexed: 12/04/2022] Open
Abstract
Transplant recipients exhibit an impaired protective immunity after SARS-CoV-2 vaccination, potentially caused by mycophenolate (MPA) immunosuppression. Recent data from patients with autoimmune disorders suggest that temporary MPA hold might greatly improve booster vaccination outcomes. We applied a fourth dose of SARS-CoV-2 vaccine to 29 kidney transplant recipients during a temporary (5 weeks) MPA/azathioprine hold, who had not mounted a humoral immune response to previous vaccinations. Seroconversion until day 32 after vaccination was observed in 76% of patients, associated with acquisition of virus-neutralizing capacity. Interestingly, 21/25 (84%) calcineurin inhibitor-treated patients responded, but only 1/4 belatacept-treated patients responded. In line with humoral responses, counts and relative frequencies of spike receptor binding domain-specific (RBD-specific) B cells were markedly increased on day 7 after vaccination, with an increase in RBD-specific CD27++CD38+ plasmablasts. Whereas overall proportions of spike-reactive CD4+ T cells remained unaltered after the fourth dose, frequencies were positively correlated with specific IgG levels. Importantly, antigen-specific proliferating Ki67+ and in vivo-activated programmed cell death 1-positive T cells significantly increased after revaccination during MPA hold, whereas cytokine production and memory differentiation remained unaffected. In summary, antimetabolite hold augmented all arms of immunity during booster vaccination. These data suggest further studies of antimetabolite hold in kidney transplant recipients.
Collapse
Affiliation(s)
- Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care and
- Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Hector Rincon-Arevalo
- Department of Nephrology and Medical Intensive Care and
- Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre Berlin (DRFZ), Berlin, Germany
- Cellular Immunology and Immunogenetics Group, Faculty of Medicine, Institute of Medical Research, University of Antioquia (UdeA), Medellín, Colombia
| | - Annika Jens
- Department of Nephrology and Medical Intensive Care and
| | - Ana-Luisa Stefanski
- Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre Berlin (DRFZ), Berlin, Germany
| | | | | | - Nadine Koch
- Department of Nephrology and Medical Intensive Care and
| | | | - Julia Beck
- Department of Clinical Pharmacology, Universitätsmedizin Göttingen, Göttingen, Germany
- Chronix Biomedical GmbH, Göttingen, Germany
| | - Michael Oellerich
- Department of Clinical Pharmacology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Vanessa Proß
- Department for General and Visceral Surgery, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carolin Stahl
- Department for General and Visceral Surgery, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care and
| | | | - Lutz Liefeldt
- Department of Nephrology and Medical Intensive Care and
| | - Petra Glander
- Department of Nephrology and Medical Intensive Care and
| | | | | | | | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg–Hessen and University Hospital Ulm, Ulm, Germany
| | - Carolin Ludwig
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg–Hessen and University Hospital Ulm, Ulm, Germany
| | - Bernd Jahrsdörfer
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg–Hessen and University Hospital Ulm, Ulm, Germany
| | | | - Nils Lachmann
- Center for Tumor Medicine, H&I Laboratory, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katja Kotsch
- Department for General and Visceral Surgery, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre Berlin (DRFZ), Berlin, Germany
| | | | - Arne Sattler
- Department for General and Visceral Surgery, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care and
| |
Collapse
|
21
|
Saharia KK, Husson JS, Niederhaus SV, Iraguha T, Avila SV, Yoo YJ, Hardy NM, Fan X, Omili D, Crane A, Carrier A, Xie WY, Vander Mause E, Hankey K, Bauman S, Lesho P, Mannuel HD, Ahuja A, Mathew M, Avruch J, Baddley J, Goloubeva O, Shetty K, Dahiya S, Rapoport AP, Luetkens T, Atanackovic D. Humoral immunity against SARS-CoV-2 variants including omicron in solid organ transplant recipients after three doses of a COVID-19 mRNA vaccine. Clin Transl Immunology 2022; 11:e1391. [PMID: 35505864 PMCID: PMC9052011 DOI: 10.1002/cti2.1391] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives Solid organ transplant recipients (SOTR) receiving post‐transplant immunosuppression show increased COVID‐19‐related mortality. It is unclear whether an additional dose of COVID‐19 vaccines can overcome the reduced immune responsiveness against severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) variants. Methods We analysed humoral immune responses against SARS‐CoV‐2 and its variants in 53 SOTR receiving SARS‐CoV‐2 vaccination. Results Following the initial vaccination series, 60.3% of SOTR showed no measurable neutralisation and only 18.9% demonstrated neutralising activity of > 90%. More intensive immunosuppression, antimetabolites in particular, negatively impacted antiviral immunity. While absolute IgG levels were lower in SOTR than controls, antibody titres against microbial recall antigens were higher. By contrast, SOTR showed reduced vaccine‐induced IgG/IgA antibody titres against SARS‐CoV‐2 and its delta variants and fewer linear B‐cell epitopes, indicating reduced B‐cell diversity. Importantly, a third vaccine dose led to an increase in anti‐SARS‐CoV‐2 antibody titres and neutralising activity across alpha, beta and delta variants and to the induction of anti‐SARS‐CoV‐2 CD4+ T cells in a subgroup of patients analysed. By contrast, we observed significantly lower antibody titres after the third dose with the omicron variant compared to the ancestral SARS‐CoV‐2 and the improvement in neutralising activity was much less pronounced than for all the other variants. Conclusion Only a small subgroup of solid organ transplant recipients is able to generate functional antibodies after an initial vaccine series; however, an additional vaccine dose resulted in dramatically improved antibody responses against all SARS‐CoV‐2 variants except omicron where antibody responses and neutralising activity remained suboptimal.
Collapse
Affiliation(s)
- Kapil K Saharia
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD USA.,Divison of Infectious Diseases University of Maryland School of Medicine Baltimore MD USA
| | - Jennifer S Husson
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD USA.,Divison of Infectious Diseases University of Maryland School of Medicine Baltimore MD USA
| | - Silke V Niederhaus
- Department of Surgery University of Maryland School of Medicine Baltimore MD USA
| | - Thierry Iraguha
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Stephanie V Avila
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Youngchae J Yoo
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD USA
| | - Nancy M Hardy
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Xiaoxuan Fan
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Destiny Omili
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Alice Crane
- Department of Surgery University of Maryland School of Medicine Baltimore MD USA
| | - Amber Carrier
- Department of Surgery University of Maryland School of Medicine Baltimore MD USA
| | - Wen Y Xie
- University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA.,Department of Surgery University of Florida College of Medicine Gainesville FL USA
| | - Erica Vander Mause
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Kim Hankey
- Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Sherri Bauman
- Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Patricia Lesho
- Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Heather D Mannuel
- University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA.,Baltimore Veterans Affairs Medical Center Baltimore MD USA
| | - Ashish Ahuja
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Minu Mathew
- Divison of Infectious Diseases University of Maryland School of Medicine Baltimore MD USA
| | - James Avruch
- Department of Surgery University of Maryland School of Medicine Baltimore MD USA
| | - John Baddley
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD USA.,Divison of Infectious Diseases University of Maryland School of Medicine Baltimore MD USA.,University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Olga Goloubeva
- Department of Epidemiology and Public Health University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Kirti Shetty
- Division of Hepatology/Liver Transplantation University of Maryland School of Medicine Baltimore MD USA
| | - Saurabh Dahiya
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Aaron P Rapoport
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Tim Luetkens
- Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA.,Department of Microbiology and Immunology University of Maryland Baltimore MD USA
| | - Djordje Atanackovic
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA.,Department of Microbiology and Immunology University of Maryland Baltimore MD USA
| |
Collapse
|
22
|
Yiu G, Rasmussen TK, Tsai BL, Diep VK, Haddon DJ, Tsoi J, Miller GD, Comin-Anduix B, Deleuran B, Crooks GM, Utz PJ. High Interferon Signature Leads to Increased STAT1/3/5 Phosphorylation in PBMCs From SLE Patients by Single Cell Mass Cytometry. Front Immunol 2022; 13:833636. [PMID: 35185925 PMCID: PMC8851522 DOI: 10.3389/fimmu.2022.833636] [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: 12/11/2021] [Accepted: 01/11/2022] [Indexed: 12/02/2022] Open
Abstract
The establishment of an “interferon (IFN) signature” to subset SLE patients on disease severity has led to therapeutics targeting IFNα. Here, we investigate IFN signaling in SLE using multiplexed protein arrays and single cell cytometry by time of flight (CyTOF). First, the IFN signature for SLE patients (n=81) from the Stanford Lupus Registry is determined using fluidigm qPCR measuring 44 previously determined IFN-inducible transcripts. IFN-high (IFN-H) patients have increased SLE criteria and renal/CNS/immunologic involvement, and increased autoantibody reactivity against spliceosome-associated antigens. CyTOF analysis is performed on non-stimulated and stimulated (IFNα, IFNγ, IL-21) PBMCs from SLE patients (n=25) and HCs (n=9) in a panel identifying changes in phosphorylation of intracellular signaling proteins (pTOF). Another panel is utilized to detect changes in intracellular cytokine (ICTOF) production in non-stimulated and stimulated (PMA/ionomycin) PBMCs from SLE patients (n=31) and HCs (n=17). Bioinformatic analysis by MetaCyto and OMIQ reveal phenotypic changes in immune cell subsets between IFN-H and IFN-low (IFN-L) patients. Most notably, IFN-H patients exhibit increased STAT1/3/5 phosphorylation downstream of cytokine stimulation and increased phosphorylation of non-canonical STAT proteins. These results suggest that IFN signaling in SLE modulates STAT phosphorylation, potentially uncovering possible targets for future therapeutic approaches.
Collapse
Affiliation(s)
- Gloria Yiu
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States.,Department of Rheumatology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tue Kruse Rasmussen
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Brandon L Tsai
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Vivian K Diep
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States
| | - David J Haddon
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States
| | - Jennifer Tsoi
- Department of Surgery David Geffen School of Medicine, Johnson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Gopika D Miller
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States
| | - Begoña Comin-Anduix
- Department of Surgery David Geffen School of Medicine, Johnson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, United States.,Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Gay M Crooks
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, United States.,Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Eli and Edythe Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles (UCLA), Los Angeles, CA, United States.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Paul J Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States.,Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
23
|
Zeidi M, Chen KL, Patel J, Desai K, Kim HJ, Chakka S, Lim R, Werth VP. Increased CD69+CCR7+ circulating activated T cells and STAT3 expression in cutaneous lupus erythematosus patients recalcitrant to antimalarials. Lupus 2022; 31:472-481. [PMID: 35258358 DOI: 10.1177/09612033221084093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Antimalarials are first-line systemic therapy for cutaneous lupus erythematosus (CLE). While some patients unresponsive to hydroxychloroquine (HCQ) alone benefit from the addition of quinacrine (QC), a subset of patients is refractory to both antimalarials. METHODS We classified CLE patients as HCQ-responders, HCQ+QC-responders, or HCQ+QC-nonresponders to compare immune profiles. Immunohistochemistry, immunofluorescence, and qRT-PCR were used to characterize inflammatory cells and cytokines in lesional skin. RESULTS Immunohistochemistry showed that CD69+ T cells were higher in HCQ+QC-nonresponders compared to HCQ- and HCQ+QC-responders (p < 0.05). Immunofluorescence further identified these cells as CD69+CCR7+ circulating activated T cells. Myeloid dendritic cells were significantly higher in HCQ+QC-responders compared to both HCQ-responders and HCQ+QC-nonresponders. Plasmacytoid dendritic cells were significantly increased in HCQ-responders compared to HCQ- and HCQ+QC-nonresponders. No differences were found in the number of autoreactive T cells, MAC387+ cells, and neutrophils among the groups. CLASI scores of the HCQ+QC-nonresponder group positively correlated with CD69+CCR7+ circulating activated T cells (r = 0.6335, p < 0.05) and MAC387+ cells (r = 0.5726, p < 0.05). IL-17 protein expression was higher in HCQ+QC-responders compared to HCQ-responders or HCQ+QC-nonresponders, while IL-22 protein expression did not differ. mRNA expression demonstrated increased STAT3 expression in a subset of HCQ+QC-nonresponders. CONCLUSION An increased number of CD69+CCR7+ circulating activated T cells and a strong correlation with CLASI scores in the HCQ+QC-nonresponders suggest these cells are involved in antimalarial-refractory skin disease. STAT3 is also increased in HCQ+QC-nonresponders and may also be a potential target for antimalarial-refractory skin disease.
Collapse
Affiliation(s)
- Majid Zeidi
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Kristen L Chen
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Jay Patel
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Krisha Desai
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Hee Joo Kim
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Srita Chakka
- Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Lim
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA
| | - Victoria P Werth
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Huang MW, Stock AD, Putterman C. CXCL13 Neutralization Attenuates Neuropsychiatric Manifestations in Lupus-Prone Mice. Front Immunol 2021; 12:763065. [PMID: 34868008 PMCID: PMC8633419 DOI: 10.3389/fimmu.2021.763065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/04/2021] [Indexed: 01/02/2023] Open
Abstract
Neuropsychiatric lupus (NPSLE), the nervous system presentation of systemic lupus erythematosus (SLE), remains challenging to treat due to its unclear pathogenesis and lack of available targeted therapies. A potential contributor to disease progression is brain tertiary lymphoid structures (TLS); these ectopic lymphoid follicles that can develop tissue-targeted antibodies have recently been described in the MRL/lpr lupus mouse strain, a classic model for studying NPSLE. The brains of MRL/lpr mice show a significant increase of CXCL13, an important chemokine in lymphoid follicle formation and retention that may also play a role in the disease progression of NPSLE. The aim of the present study was to inhibit CXCL13 and examine the effect of this intervention on lymphoid formation and the development of neurobehavioral manifestations in lupus mice. Female MRL/lpr mice were injected with an anti-CXCL13 antibody, an IgG1 isotype-matched antibody, or PBS either three times a week for 12 weeks intraperitoneally (IP) starting at 6-8 weeks of age, or continuously intracerebroventricularly (ICV) with an osmotic pump over a two-week period starting at 15 weeks of age. Cognitive dysfunction and depression-like behavior were assessed at the end of treatment. When treatment was delivered IP, anti-CXCL13 treated mice showed significant improvement in cognitive function when compared to control treated mice. Depression-like behavior was attenuated as well. Furthermore, mice that received anti-CXCL13 by the ICV route showed similar beneficial effects. However, the extent of lymphocyte infiltration into the brain and the general composition of the aggregates were not substantively changed by anti-CXCL13 irrespective of the mode of administration. Nevertheless, analysis of brain gene expression in anti-CXCL13 treated mice showed significant differences in key immunological and neuro-inflammatory pathways that most likely explained the improvement in the behavioral phenotype. Our results indicate that CXCL13 affects the behavioral manifestations in the MRL/lpr strain and is important to the pathogenesis of murine NPSLE, suggesting it as a potential therapeutic target.
Collapse
Affiliation(s)
- Michelle W Huang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ariel D Stock
- Department of Neurological Surgery, Montefiore Medical Center, Bronx, NY, United States
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, United States.,Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel.,Galilee Medical Center Research Institute, Nahariya, Israel
| |
Collapse
|
25
|
Paquissi FC, Abensur H. The Th17/IL-17 Axis and Kidney Diseases, With Focus on Lupus Nephritis. Front Med (Lausanne) 2021; 8:654912. [PMID: 34540858 PMCID: PMC8446428 DOI: 10.3389/fmed.2021.654912] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/04/2021] [Indexed: 12/28/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a disease characterized by dysregulation and hyperreactivity of the immune response at various levels, including hyperactivation of effector cell subtypes, autoantibodies production, immune complex formation, and deposition in tissues. The consequences of hyperreactivity to the self are systemic and local inflammation and tissue damage in multiple organs. Lupus nephritis (LN) is one of the most worrying manifestations of SLE, and most patients have this involvement at some point in the course of the disease. Among the effector cells involved, the Th17, a subtype of T helper cells (CD4+), has shown significant hyperactivation and participates in kidney damage and many other organs. Th17 cells have IL-17A and IL-17F as main cytokines with receptors expressed in most renal cells, being involved in the activation of many proinflammatory and profibrotic pathways. The Th17/IL-17 axis promotes and maintains repetitive tissue damage and maladaptive repair; leading to fibrosis, loss of organ architecture and function. In the podocytes, the Th17/IL-17 axis effects include changes of the cytoskeleton with increased motility, decreased expression of health proteins, increased oxidative stress, and activation of the inflammasome and caspases resulting in podocytes apoptosis. In renal tubular epithelial cells, the Th17/IL-17 axis promotes the activation of profibrotic pathways such as increased TGF-β expression and epithelial-mesenchymal transition (EMT) with consequent increase of extracellular matrix proteins. In addition, the IL-17 promotes a proinflammatory environment by stimulating the synthesis of inflammatory cytokines by intrinsic renal cells and immune cells, and the synthesis of growth factors and chemokines, which together result in granulopoiesis/myelopoiesis, and further recruitment of immune cells to the kidney. The purpose of this work is to present the prognostic and immunopathologic role of the Th17/IL-17 axis in Kidney diseases, with a special focus on LN, including its exploration as a potential immunotherapeutic target in this complication.
Collapse
Affiliation(s)
- Feliciano Chanana Paquissi
- Department of Medicine, Clínica Girassol, Luanda, Angola
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Hugo Abensur
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Ghafouri-Fard S, Shahir M, Taheri M, Salimi A. A review on the role of chemokines in the pathogenesis of systemic lupus erythematosus. Cytokine 2021; 146:155640. [PMID: 34252872 DOI: 10.1016/j.cyto.2021.155640] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022]
Abstract
Chemokines are a group of cytokines with low molecular weight that principally direct chemotaxis of target cells. They have prominent roles in the pathogenesis systemic lupus erythematosus (SLE) and related complications particularly lupus nephritis. These molecules not only induce autoimmune responses in the organs of patients, but also can amplify the induced inflammatory responses. Although chemokine family has at least 46 identified members, the role of a number of these molecules have been more clarified in SLE patients or animal models of this disorder. In the current paper, we review the role of CCL2, CCL3, CCL4, CCL11, CCL20, CXCL1, CXCL2, CXCL8, CXCL10, CXCL12 and CXCL13 in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehri Shahir
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Alireza Salimi
- Critical Care Quality Improvement Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Vascular Endothelial Growth Factor Biology and Its Potential as a Therapeutic Target in Rheumatic Diseases. Int J Mol Sci 2021; 22:ijms22105387. [PMID: 34065409 PMCID: PMC8161097 DOI: 10.3390/ijms22105387] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Rheumatic diseases constitute a diversified group of diseases distinguished by arthritis and often involve other organs. The affected individual has low quality of life, productivity even life-threatening in some severe conditions. Moreover, they impose significant economic and social burdens. In recent years, the patient outcome has been improved significantly due to clearer comprehension of the pathology of rheumatic diseases and the effectiveness of "treat to target" therapies. However, the high cost and the adverse effects are the concerns and full remissions are not often observed. One of the main processes that contributes to the pathogenesis of rheumatic diseases is angiogenesis. Vascular endothelial growth factor (VEGF), a central mediator that regulates angiogenesis, has different isoforms and functions in various physiological processes. Increasing evidence suggests an association between the VEGF system and rheumatic diseases. Anti-VEGF and VEGF receptor (VEGFR) therapies have been used to treat several cancers and eye diseases. This review summarizes the current understanding of VEGF biology and its role in the context of rheumatic diseases, the contribution of VEGF bioavailability in the pathogenesis of rheumatic diseases, and the potential implications of therapeutic approaches targeting VEGF for these diseases.
Collapse
|
28
|
Robles-Vera I, de la Visitación N, Toral M, Sánchez M, Gómez-Guzmán M, Jiménez R, Romero M, Duarte J. Mycophenolate mediated remodeling of gut microbiota and improvement of gut-brain axis in spontaneously hypertensive rats. Biomed Pharmacother 2021; 135:111189. [PMID: 33388596 DOI: 10.1016/j.biopha.2020.111189] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/27/2020] [Accepted: 12/26/2020] [Indexed: 12/15/2022] Open
Abstract
Microbiota has a role in the host blood pressure (BP) regulation. The immunosuppressive drug mofetil mycophenolate (MMF) ameliorates hypertension. The present study analyzes whether MMF improves dysbiosis in a genetic model of hypertension. Twenty weeks old male spontaneously hypertensive rats (SHR) and Wistar Kyoto rats (WKY) were randomly divided into three groups: untreated WKY, untreated SHR, and SHR treated with MMF for 5 weeks. MMF treatment restored gut bacteria from the phyla Firmicutes and Bacteroidetes, and acetate- and lactate-producing bacteria to levels similar to those found in WKY, increasing butyrate-producing bacteria. MMF increased the percentage of anaerobic bacteria in the gut. The improvement of gut dysbiosis was associated with an enhanced colonic integrity and a decreased sympathetic drive in the gut. MMF inhibited neuroinflammation in the paraventricular nuclei in the hypothalamus. MMF increased the lower regulatory T cells proportion in mesenteric lymph nodes and Th17 and Th1 infiltration in aorta, improved aortic endothelial function and reduced systolic BP. This study demonstrates for the first time that MMF reduces gut dysbiosis in SHR. This effect could be related to its capability to improve gut integrity due to reduced sympathetic drive in the gut associated to the reduced brain neuroinflammation.
Collapse
Affiliation(s)
- Iñaki Robles-Vera
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain
| | - Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain
| | - Marta Toral
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain; Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Manuel Sánchez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Manuel Gómez-Guzmán
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain.
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain.
| |
Collapse
|
29
|
Klotsman M, Coquery S, Sathyan G, Naageshwaran V, Shivanand P, Fairchild AJ, Garden OA, Anderson WH. Pharmacokinetics and Pharmacodynamics of Immediate- and Modified-Release Mycophenolic Acid Preparations in Healthy Beagle Dogs. Front Vet Sci 2021; 7:611404. [PMID: 33585601 PMCID: PMC7876310 DOI: 10.3389/fvets.2020.611404] [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: 10/28/2020] [Accepted: 12/22/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Mycophenolic acid (MPA) is a broad-acting immunomodulating agent that may be therapeutically beneficial for the treatment of immune-mediated diseases in canine patients. Objectives: To determine the suppressive effects of MPA on T-cell proliferation, and to assess the feasibility of a canine-specific q24 h modified-release MPA formulation (OKV-1001b). Animals: Fifteen healthy purpose-bred male beagle dogs. Methods: Two nearly identical open-label fifteen-day studies were conducted in which dogs were randomized to receive mycophenolate mofetil (MMF; 10 mg/kg q12h), or two doses of OKV-1001b (270 mg and 180 mg; q24h). Serial pharmacokinetic (PK) and pharmacodynamic (PD) samples were collected on Days 1, 8, and 15. MPA plasma concentrations were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS), while an ex vivo T-cell proliferation assay assessed PD effects. Dogs were continuously monitored for evidence of side effects and gastrointestinal tolerability. Results: MPA induced inhibition of T-cell proliferation was observed following administration of all MPA preparations in a clear concentration-dependent manner. The PK/PD relationship was maintained across all days and time-points. Data generated herein suggest that MPA plasma concentrations above 600 ng/mL achieve at least 50% inhibition of T-cell proliferation. Conclusions and Clinical Importance: MPA holds therapeutic potential for treating dogs with immune-mediated disease, but clinical trials will be necessary to determine its safety and efficacy in naturally occurring disease. Likewise, q24h oral modified release MPA preparations that maintain MPA plasma concentrations between 600 and 1,000 ng/mL are warranted for further studies in client-owned dogs.
Collapse
Affiliation(s)
| | | | | | | | | | - Amanda J Fairchild
- Department of Psychology, University of South Carolina, Columbia, SC, United States
| | - Oliver A Garden
- Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States
| | - Wayne H Anderson
- Okava Pharmaceuticals, San Francisco, CA, United States.,Pulmonary and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
30
|
Slight-Webb S, Smith M, Bylinska A, Macwana S, Guthridge C, Lu R, Merrill JT, Chakravarty E, Arriens C, Munroe ME, Maecker HT, Utz PJ, Guthridge JM, James JA. Autoantibody-positive healthy individuals with lower lupus risk display a unique immune endotype. J Allergy Clin Immunol 2020; 146:1419-1433. [PMID: 32446964 PMCID: PMC7680268 DOI: 10.1016/j.jaci.2020.04.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autoimmune diseases comprise a spectrum of illnesses and are on the rise worldwide. Although antinuclear antibodies (ANAs) are detected in many autoimmune diseases, up to 20% of healthy women are ANA-positive (ANA+) and most will never develop clinical symptoms. Furthermore, disease transition is higher among ANA+ African Americans compared with ANA+ European Americans. OBJECTIVE We sought to determine the immune features that might define and prevent transition to clinical autoimmunity in ANA+ healthy individuals. METHODS We comprehensively phenotyped immune profiles of African Americans and European Americans who are ANA-negative (ANA-) healthy, ANA+ healthy, or have SLE using single cell mass cytometry, next-generation RNA-sequencing, multiplex cytokine profiling, and phospho-signaling analyses. RESULTS We found that, compared with both ANA- and ANA+ healthy individuals, patients with SLE of both races displayed T-cell expansion and elevated expression of type I and II interferon pathways. We discovered a unique immune signature that suggests a suppressive immune phenotype and reduced CD11C+ autoimmunity-associated B cells in healthy ANA+ European Americans that is absent in their SLE or even healthy ANA- counterparts, or among African American cohorts. In contrast, ANA+ healthy African Americans exhibited elevated expression of T-cell activation markers and higher plasma levels of IL-6 than did healthy ANA+ European Americans. CONCLUSIONS We propose that this novel immune signature identified in ANA+ healthy European Americans may protect them from T-cell expansion, heightened activation of interferon pathways, and disease transition.
Collapse
Affiliation(s)
- Samantha Slight-Webb
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla
| | - Miles Smith
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla
| | - Aleksandra Bylinska
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla
| | - Susan Macwana
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla
| | - Carla Guthridge
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla
| | - Rufei Lu
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Okla
| | - Joan T Merrill
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla
| | - Eliza Chakravarty
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla
| | - Cristina Arriens
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla; Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Okla; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Okla
| | - Melissa E Munroe
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla
| | - Holden T Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Paul J Utz
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, Calif
| | - Joel M Guthridge
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Okla
| | - Judith A James
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Okla; Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Okla; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Okla.
| |
Collapse
|
31
|
Cytokines and Transgenic Matrix in Autoimmune Diseases: Similarities and Differences. Biomedicines 2020; 8:biomedicines8120559. [PMID: 33271810 PMCID: PMC7761121 DOI: 10.3390/biomedicines8120559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases are increasingly recognized as disease entities in which dysregulated cytokines contribute to tissue-specific inflammation. In organ-specific and multiorgan autoimmune diseases, the cytokine profiles show some similarities. Despite these similarities, the cytokines have different roles in the pathogenesis of different diseases. Altered levels or action of cytokines can result from changes in cell signaling. This article describes alterations in the JAK-STAT, TGF-β and NF-κB signaling pathways, which are involved in the pathogenesis of multiple sclerosis and systemic lupus erythematosus. There is a special focus on T cells in preclinical models and in patients afflicted with these chronic inflammatory diseases.
Collapse
|
32
|
Zhang T, Warden AR, Li Y, Ding X. Progress and applications of mass cytometry in sketching immune landscapes. Clin Transl Med 2020; 10:e206. [PMID: 33135337 PMCID: PMC7556381 DOI: 10.1002/ctm2.206] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Recently emerged mass cytometry (cytometry by time-of-flight [CyTOF]) technology permits the identification and quantification of inherently diverse cellular systems, and the simultaneous measurement of functional attributes at the single-cell resolution. By virtue of its multiplex ability with limited need for compensation, CyTOF has led a critical role in immunological research fields. Here, we present an overview of CyTOF, including the introduction of CyTOF principle and advantages that make it a standalone tool in deciphering immune mysteries. We then discuss the functional assays, introduce the bioinformatics to interpret the data yield via CyTOF, and depict the emerging clinical and research applications of CyTOF technology in sketching immune landscape in a wide variety of diseases.
Collapse
Affiliation(s)
- Ting Zhang
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Antony R. Warden
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yiyang Li
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Xianting Ding
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
33
|
Moore E, Putterman C. Are lupus animal models useful for understanding and developing new therapies for human SLE? J Autoimmun 2020; 112:102490. [PMID: 32535128 PMCID: PMC7384952 DOI: 10.1016/j.jaut.2020.102490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023]
Abstract
Systemic lupus erythematosus is a systemic autoimmune disease driven by a complex combination of genetic, environmental, and other immunoregulatory factors. The development of targeted therapies is complicated by heterogeneous clinical manifestations, varying organ involvement, and toxicity. Despite advances in understanding the mechanisms contributing to SLE, only one biologic drug, belimumab, is FDA-approved. The identification and development of potential therapies have largely been driven by studies in lupus animal models. Therefore, direct comparison of both the therapeutic and immunological findings in human and murine SLE studies is critical and can reveal important insights into indeed how useful and relevant are murine studies in SLE drug development. Studies involving belimumab, mycophenolate mofetil, abatacept, rituximab, and anti-interferon strategies generally demonstrated analogous findings in the attenuation of SLE manifestations and modulation of select immune cell populations in human and murine SLE. While further basic and translational studies are needed to identify SLE patient subsets likely to respond to particular therapeutic modalities and in dissecting complex mechanisms, we believe that despite some inherent weaknesses SLE mouse models will continue to be integral in developing targeted SLE therapies.
Collapse
Affiliation(s)
- Erica Moore
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA; Bar-Ilan University Azrieli Faculty of Medicine, Safed, Israel; Research Institute, Galilee Medical Center, Nahariya, Israel.
| |
Collapse
|
34
|
Mycophenolate mofetil attenuates concanavalin A-induced acute liver injury through modulation of TLR4/NF-κB and Nrf2/HO-1 pathways. Pharmacol Rep 2020; 72:945-955. [PMID: 32048261 DOI: 10.1007/s43440-019-00055-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/24/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Acute liver injury (ALI) is a serious health condition associated with rising morbidity and sudden progression. This study was designed to investigate the possible hepatocurative potential of two dose levels (30 and 60 mg/kg) of Mycophenolate mofetil (MMF), an immune-suppressant agent, against Concanavalin A (Con A)-induced ALI in mice. METHOD A single dose of Con A (20 mg/kg, IV) was used to induce ALI in mice. MMF (30 mg/kg and 60 mg/kg) was administered orally for 4 days post Con A injection. RESULTS MMF (30 mg/kg) failed to cause significant amelioration in Con A-induced ALI while MMF (60 mg/kg) significantly alleviated Con A-induced ALI. Administration of MMF (60 mg/kg) significantly decreased Con A-induced increase in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. Additionally, MMF significantly restored the disrupted oxidant/antioxidants status induced by Con A. MMF caused marked increase in hepatic nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) levels. Moreover, MMF significantly reduced Con A-induced increase in the expression of hepatic toll-like receptor 4 (TLR4), nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α), interferon-γ (INF-γ) and interleukin-1β (Il-1β). Also, MMF administration significantly decreased Con A-induced increase in the immune-expression of pro-apoptotic Bcl-2-associated X protein (Bax) and markedly increased Con A-induced decrease in the anti-apoptotic B-cell lymphoma 2 protein (Bcl2). CONCLUSION The observed ameliorative effect of MMF against Con A-induce ALI may be contributed to its anti-inflammatory, anti-oxidant and anti-apoptotic potentials taking into consideration that TLR4/NF-κB and Nrf2/HO-1 are the main implicated pathways. Schematic diagram summarizing the possible mechanisms underlying the ameliorative potential of Mycophenolate Mofetil against Con A-induced acute liver injury. Bax Bcl-2-associated X protein, Bcl2 B-cell lymphoma 2, MMF Mycophenolate mofetil, Con A Concanavalin A, GSH reduced glutathione, HO-1 Heme oxygenase-1, IL-1β Interleukin-1β, IFN-γ Interferon-γ, MDA Malondialdehyde, NF-κB Nuclear Factor Kappa B, Nrf2 Nuclear factor erythroid 2-related factor 2, NO Nitric Oxide, SOD Superoxide Dismutase, TLR4 Toll-like receptor 4, TNF-α tumor necrosis factor-α.
Collapse
|
35
|
Logotheti S, Pützer BM. STAT3 and STAT5 Targeting for Simultaneous Management of Melanoma and Autoimmune Diseases. Cancers (Basel) 2019; 11:cancers11101448. [PMID: 31569642 PMCID: PMC6826843 DOI: 10.3390/cancers11101448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/16/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
Melanoma is a skin cancer which can become metastatic, drug-refractory, and lethal if managed late or inappropriately. An increasing number of melanoma patients exhibits autoimmune diseases, either as pre-existing conditions or as sequelae of immune-based anti-melanoma therapies, which complicate patient management and raise the need for more personalized treatments. STAT3 and/or STAT5 cascades are commonly activated during melanoma progression and mediate the metastatic effects of key oncogenic factors. Deactivation of these cascades enhances antitumor-immune responses, is efficient against metastatic melanoma in the preclinical setting and emerges as a promising targeting strategy, especially for patients resistant to immunotherapies. In the light of the recent realization that cancer and autoimmune diseases share common mechanisms of immune dysregulation, we suggest that the systemic delivery of STAT3 or STAT5 inhibitors could simultaneously target both, melanoma and associated autoimmune diseases, thereby decreasing the overall disease burden and improving quality of life of this patient subpopulation. Herein, we review the recent advances of STAT3 and STAT5 targeting in melanoma, explore which autoimmune diseases are causatively linked to STAT3 and/or STAT5 signaling, and propose that these patients may particularly benefit from treatment with STAT3/STAT5 inhibitors.
Collapse
Affiliation(s)
- Stella Logotheti
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany.
- Department Life, Light & Matter, University of Rostock, 18059 Rostock, Germany.
| |
Collapse
|