1
|
Meng X, Zhong Y, Kuang X, Zhang Y, Yang L, Cai Y, Wang F, He F, Xie H, Wang B, Li J. Targeting the STAT3/IL-36G signaling pathway can be a promising approach to treat rosacea. J Adv Res 2025; 71:429-440. [PMID: 38909883 DOI: 10.1016/j.jare.2024.06.013] [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: 03/06/2024] [Revised: 05/26/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Rosacea is an inflammatory skin disorder characterized by the release of inflammatory mediators from keratinocytes, which are thought to play a crucial role in its pathogenesis. Despite an incidence of approximately 5.5%, rosacea is associated with a poor quality of life. However, as the pathogenesis of rosacea remains enigmatic, treatment options are limited. OBJECTIVES To investigate the pathogenesis of rosacea and explore new therapeutic strategies. METHODS Transcriptome data from rosacea patients combined with immunohistochemical staining were used to investigate the activation of STAT3 in rosacea. The role of STAT3 activation in rosacea was subsequently explored by inhibiting STAT3 activation both in vivo and in vitro. The key molecules downstream of STAT3 activation were identified through data analysis and experiments. Dual-luciferase assay and ChIP-qPCR analysis were used to validate the direct binding of STAT3 to the IL-36G promoter. DARTS, in combination with experimental screening, was employed to identify effective drugs targeting STAT3 for rosacea treatment. RESULTS STAT3 signaling was hyperactivated in rosacea and served as a promoter of the keratinocyte-driven inflammatory response. Mechanistically, activated STAT3 directly bind to the IL-36G promoter region to amplify downstream inflammatory signals by promoting IL-36G transcription, and treatment with a neutralizing antibody (α-IL36γ) could mitigate rosacea-like inflammation. Notably, a natural plant extract (pogostone), which can interact with STAT3 directly to inhibit its activation and affect the STAT3/IL36G signaling pathway, was screened as a promising topical medication for rosacea treatment. CONCLUSIONS Our study revealed a pivotal role for STAT3/IL36G signaling in the development of rosacea, suggesting that targeting this pathway might be a potential strategy for rosacea treatment.
Collapse
Affiliation(s)
- Xin Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Yun Zhong
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuyuan Kuang
- Department of Hyperbaric Oxygen, Xiangya Hospital, Central South University, Jiangxi, China; Department of Hyperbaric Oxygen, Xiangya Hospital, Central South University, Changsha, China
| | - Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Li Yang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yisheng Cai
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Fan Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Fanping He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; Department of Plastic and Reconstructive Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Ben Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Samad MA, Ahmad I, Hasan A, Alhashmi MH, Ayub A, Al‐Abbasi FA, Kumer A, Tabrez S. STAT3 Signaling Pathway in Health and Disease. MedComm (Beijing) 2025; 6:e70152. [PMID: 40166646 PMCID: PMC11955304 DOI: 10.1002/mco2.70152] [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: 09/30/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a critical transcription factor involved in multiple physiological and pathological processes. While STAT3 plays an essential role in homeostasis, its persistent activation has been implicated in the pathogenesis of various diseases, particularly cancer, bone-related diseases, autoimmune disorders, inflammatory diseases, cardiovascular diseases, and neurodegenerative conditions. The interleukin-6/Janus kinase (JAK)/STAT3 signaling axis is central to STAT3 activation, influencing tumor microenvironment remodeling, angiogenesis, immune evasion, and therapy resistance. Despite extensive research, the precise mechanisms underlying dysregulated STAT3 signaling in disease progression remain incompletely understood, and no United States Food and Drug Administration (USFDA)-approved direct STAT3 inhibitors currently exist. This review provides a comprehensive evaluation of STAT3's role in health and disease, emphasizing its involvement in cancer stem cell maintenance, metastasis, inflammation, and drug resistance. We systematically discuss therapeutic strategies, including JAK inhibitors (tofacitinib, ruxolitinib), Src Homology 2 domain inhibitors (S3I-201, STATTIC), antisense oligonucleotides (AZD9150), and nanomedicine-based drug delivery systems, which enhance specificity and bioavailability while reducing toxicity. By integrating molecular mechanisms, disease pathology, and emerging therapeutic interventions, this review fills a critical knowledge gap in STAT3-targeted therapy. Our insights into STAT3 signaling crosstalk, epigenetic regulation, and resistance mechanisms offer a foundation for developing next-generation STAT3 inhibitors with greater clinical efficacy and translational potential.
Collapse
Affiliation(s)
- Md Abdus Samad
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Iftikhar Ahmad
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Aakifah Hasan
- Department of BiochemistryFaculty of Life ScienceAligarh Muslim UniversityAligarhIndia
| | - Mohammad Hassan Alhashmi
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
- Department of Medical Laboratory SciencesFaculty of Applied Medical SciencesKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Arusha Ayub
- Department of MedicineCollege of Health SciencesUniversity of GeorgiaGeorgiaUSA
| | - Fahad A. Al‐Abbasi
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Ajoy Kumer
- Department of ChemistryCollege of Arts and SciencesInternational University of Business Agriculture & Technology (IUBAT)DhakaBangladesh
| | - Shams Tabrez
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
- Department of Medical Laboratory SciencesFaculty of Applied Medical SciencesKing Abdulaziz UniversityJeddahSaudi Arabia
| |
Collapse
|
3
|
Metwally H. STAT Signature Dish: Serving Immunity with a Side of Dietary Control. Biomolecules 2025; 15:487. [PMID: 40305224 PMCID: PMC12024614 DOI: 10.3390/biom15040487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Immunity is a fundamental aspect of animal biology, defined as the host's ability to detect and defend against harmful pathogens and toxic substances to preserve homeostasis. However, immune defenses are metabolically demanding, requiring the efficient allocation of limited resources to balance immune function with other physiological and developmental needs. To achieve this balance, organisms have evolved sophisticated signaling networks that enable precise, context-specific responses to internal and external cues. These networks are essential for survival and adaptation in multicellular systems. Central to this regulatory architecture is the STAT (signal transducer and activator of Transcription) family, a group of versatile signaling molecules that govern a wide array of biological processes across eukaryotes. STAT signaling demonstrates remarkable plasticity, from orchestrating host defense mechanisms to regulating dietary metabolism. Despite its critical role, the cell-specific and context-dependent nuances of STAT signaling remain incompletely understood, highlighting a significant gap in our understanding. This review delves into emerging perspectives on immunity, presenting dynamic frameworks to explore the complexity and adaptability of STAT signaling and the underlying logic driving cellular decision-making. It emphasizes how STAT pathways integrate diverse physiological processes, from immune responses to dietary regulation, ultimately supporting organismal balance and homeostasis.
Collapse
Affiliation(s)
- Hozaifa Metwally
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
4
|
Zhang C, Hu S, Yin C, Wang G, Liu P. STAT3 orchestrates immune dynamics in hepatocellular carcinoma: A pivotal nexus in tumor progression. Crit Rev Oncol Hematol 2025; 207:104620. [PMID: 39818308 DOI: 10.1016/j.critrevonc.2025.104620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/04/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) presents a formidable challenge in oncology, attributed to its association with chronic liver diseases and global prevalence. The immune microenvironment profoundly influences HCC progression, balancing immune suppression and antitumor responses. The Signal Transducer and Activator of Transcription 3 (STAT3) is central to this equilibrium, orchestrating immune dynamics and intertwining tumor progression with immune evasion mechanisms. Dysregulated STAT3 signaling, activated by various stimuli, including cytokines and growth factors, promotes an immunosuppressive milieu within HCC tumors, fostering tumor survival and proliferation while hindering immune surveillance. Non-coding RNAs and other molecules regulate this process, modulating STAT3 activity and influencing immune cell function. Moreover, therapeutic interventions targeting the STAT3 pathway, alongside advancements in radiotherapy, cancer vaccines, and diabetes-related drugs, offer promising strategies in HCC management. Integrating natural compounds with immunotherapy emerges as a novel approach, leveraging their ability to enhance antitumor immunity and counter immune evasion strategies. Understanding the multifaceted role of STAT3 and its interactions within the immune landscape of HCC is paramount for devising effective therapeutic interventions and improving patient outcomes.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Songbai Hu
- Department of Cancer Center, Yuexi County Hospital, Anqing, Anhui Province 246600, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Hubei, China.
| |
Collapse
|
5
|
Ciurus S, Elewa MAF, Palmer MA, Wolf A, Hector M, Fuhrmann DC, Thomas D, Gurke R, Schwalm MP, Berger L, Zech TJ, Burgers LD, Marschalek R, Geisslinger G, Knapp S, Langmann T, Bracher F, Weigert A, Fürst R. Inhibition of DYRK1B BY C81 impedes inflammatory processes in leukocytes by reducing STAT3 activity. Cell Mol Life Sci 2025; 82:85. [PMID: 39985685 PMCID: PMC11846820 DOI: 10.1007/s00018-025-05579-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 10/15/2024] [Accepted: 01/05/2025] [Indexed: 02/24/2025]
Abstract
Chronic inflammatory diseases are a significant global burden and are associated with dysregulated resolution of inflammation. Therefore, promoting the process of resolution is a promising therapeutic approach. This study presents the potent anti-inflammatory and pro-resolving effects of a natural product-derived compound called C81. Administration of C81 in a therapeutic window resolved inflammation in the murine imiquimod-induced psoriasis model, and reduced microglial infiltration in a laser-induced choroidal neovascularisation model. Investigations into the underlying mechanisms of C81 identified the DYRK1B/STAT3 axis as a new regulator of inflammatory processes in leukocytes. The inhibition of DYRK1B by C81 resulted in attenuated STAT3 phosphorylation. The depletion of STAT3-regulated gene expression led to the inhibition of leukocyte adhesion and migration due to reduced integrin activation, and in addition to the inhibition of the release of pro-inflammatory mediators such as cytokines and eicosanoids. Importantly, the pro-resolving effects of C81 included the cell type-specific induction of apoptosis in neutrophils and a subsequent increase in efferocytosis. In conclusion, we report the DYRK1B/STAT3 axis as a novel and promising therapeutic target for activating the resolution of inflammation.
Collapse
Affiliation(s)
- Sarah Ciurus
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Mohammed A F Elewa
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
- Department of Biochemistry, Faculty of Pharmacy, Kafr El-Sheikh University, Karf El-Sheikh, Egypt
| | - Megan A Palmer
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mandy Hector
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Martin P Schwalm
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Lena Berger
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas J Zech
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
- Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Luisa D Burgers
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Franz Bracher
- Pharmaceutical Chemistry, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany.
- Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
6
|
Mackie J, Suan D, McNaughton P, Haerynck F, O’Sullivan M, Guerin A, Ma CS, Tangye SG. Functional validation of a novel STAT3 'variant of unknown significance' identifies a new case of STAT3 GOF syndrome and reveals broad immune cell defects. Clin Exp Immunol 2025; 219:uxaf005. [PMID: 39836489 PMCID: PMC11791529 DOI: 10.1093/cei/uxaf005] [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: 10/21/2024] [Revised: 11/29/2024] [Accepted: 01/20/2025] [Indexed: 01/23/2025] Open
Abstract
INTRODUCTION Signal transducer and activator of transcription 3 (STAT3) orchestrates crucial immune responses through its pleiotropic functions as a transcription factor. Patients with germline monoallelic dominant negative or hypermorphic STAT3 variants, who present with immunodeficiency and/or immune dysregulation, have revealed the importance of balanced STAT3 signaling in lymphocyte differentiation and function, and immune homeostasis. Here, we report a novel missense variant of unknown significance in the DNA-binding domain of STAT3 in a patient who experienced hypogammaglobulinemia, lymphadenopathy, hepatosplenomegaly, immune thrombocytopenia, eczema, and enteropathy over a 35-year period. METHODS In vitro demonstration of prolonged STAT3 activation due to delayed dephosphorylation, and enhanced transcriptional activity, confirmed this to be a novel pathogenic STAT3 gain-of-function variant. Peripheral blood lymphocytes from this patient, and patients with confirmed STAT3 Gain-of-function Syndrome, were collected to investigate mechanisms of disease pathogenesis. RESULTS B cell dysregulation was evidenced by a loss of class-switched memory B cells and a significantly expanded CD19hiCD21lo B cell population, likely influenced by a skewed CXCR3+ TFH population. Interestingly, unlike STAT3 dominant negative variants, cytokine secretion by activated peripheral blood STAT3 GOF CD4+ T cells and frequencies of Treg cells were intact, suggesting CD4+ T cell dysregulation likely occurs at sites of disease rather than the periphery. CONCLUSION This study provides an in-depth case study in confirming a STAT3 gain-of-function variant and identifies lymphocyte dysregulation in the peripheral blood of patients with STAT3 gain-of-function syndrome. Identifying cellular biomarkers of disease provides a flow cytometric-based screen to guide validation of additional novel STAT3 gain-of-function variants as well as provide insights into putative mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Joseph Mackie
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Daniel Suan
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Darlinghurst, NSW, Australia
| | - Peter McNaughton
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Darlinghurst, NSW, Australia
- Queensland Paediatric Immunology and Allergy Service, Queensland Children’s Hospital, South Brisbane, Australia
| | - Filomeen Haerynck
- Department of Pediatric Pulmonology, Infectious Diseases and Immunology, Ghent University Hospital, Ghent, Belgium
- Primary Immunodeficiency Research Lab, Centre for Primary Immunodeficiency Ghent, Ghent University Hospital, Ghent, Belgium
| | - Michael O’Sullivan
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Darlinghurst, NSW, Australia
- Department of Clinical Immunology and PathWest, Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Antoine Guerin
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Darlinghurst, NSW, Australia
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Darlinghurst, NSW, Australia
| |
Collapse
|
7
|
Rallabandi HR, Singh MK, Looger LL, Nath SK. Defining Mechanistic Links Between the Non-Coding Variant rs17673553 in CLEC16A and Lupus Susceptibility. Int J Mol Sci 2025; 26:314. [PMID: 39796169 PMCID: PMC11720054 DOI: 10.3390/ijms26010314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by widespread inflammation and autoantibody production. Its development and progression involve genetic, epigenetic, and environmental factors. Although genome-wide association studies (GWAS) have repeatedly identified a susceptibility signal at 16p13, its fine-scale source and its functional and mechanistic role in SLE remain unclear. We used bioinformatics to prioritize likely functional variants and validated the top candidate through various experimental techniques, including clustered regularly interspaced short palindromic repeats (CRISPR)-based genome editing in B cells. To assess the functional impact of the proposed causal variant in C-type lectin domain family 16, member A (CLEC16A), we compared autophagy levels between wild-type (WT) and knock-out (KO) cells. Systematic bioinformatics analysis identified the highly conserved non-coding intronic variant rs17673553, with the risk allele apparently affecting enhancer function and regulating several target genes, including CLEC16A itself. Luciferase reporter assays followed by chromatin immunoprecipitation-quantitative polymerase chain reaction (ChIP-qPCR) validated this enhancer activity, demonstrating that the risk allele increases the binding of enhancer histone marks (H3K27ac and H3K4me1), the CTCF-binding factor, and key immune transcription factors (GATA3 and STAT3). Knock-down of GATA3 and STAT3 via siRNA led to a significant decrease in CLEC16A expression. These regulatory effects on the target gene were further confirmed using CRISPR-based genome editing and CRISPR-dCas9-based epigenetic activation/silencing. Functionally, WT cells exhibited higher levels of starvation-induced autophagy compared to KO cells, highlighting the role of CLEC16A and the rs17673553 locus in autophagy regulation. These findings suggest that the rs17673553 locus-particularly the risk allele-drives significant allele-specific chromatin modifications and binding of multiple transcription factors, thereby mechanistically regulating the expression of target autophagy-associated genes, including CLEC16A itself. This mechanism could potentially explain the association between rs17673553 and SLE, and could underlie the signal at 16p13.
Collapse
Affiliation(s)
- Harikrishna Reddy Rallabandi
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (H.R.R.); (M.K.S.)
| | - Manish Kumar Singh
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (H.R.R.); (M.K.S.)
| | - Loren L. Looger
- Department of Neurosciences, University of California, La Jolla, San Diego, CA 92093, USA;
- Howard Hughes Medical Institute, University of California, La Jolla, San Diego, CA 92093, USA
| | - Swapan K. Nath
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (H.R.R.); (M.K.S.)
| |
Collapse
|
8
|
Almouh M, Soukkarieh C, Kassouha M, Ibrahim S. Crosstalk between circular RNAs and the STAT3 signaling pathway in human cancer. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195051. [PMID: 39121909 DOI: 10.1016/j.bbagrm.2024.195051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Circular RNAs (circRNAs) are endogenous covalently closed single-stranded RNAs produced by reverse splicing of pre-mRNA. Emerging evidence suggests that circRNAs contribute to cancer progression by modulating the oncogenic STAT3 signaling pathway, which plays key roles in human malignancies. STAT3 signaling-related circRNAs expression appears to be extensively dysregulated in diverse cancer types, where they function either as tumor suppressors or oncogenes. However, the biological effects of STAT3 signaling-related circRNAs and their associations with cancer have not been systematically studied before. Given this, shedding light on the interaction between circRNAs and STAT3 signaling pathway in human malignancies may provide several novel insights into cancer therapy. In this review, we provide a comprehensive introduction to the molecular mechanisms by which circRNAs regulate STAT3 signaling in cancer progression, and the crosstalk between STAT3 signaling-related circRNAs and other signaling pathways. We also further discuss the role of the circRNA/STAT3 axis in cancer chemotherapy sensitivity.
Collapse
Affiliation(s)
- Mansour Almouh
- Department of Animal Production, Faculty of Veterinary Medicine, Hama University, Hama, Syria.
| | - Chadi Soukkarieh
- Department of Animal Biology, Faculty of Sciences, Damascus University, Damascus, Syria
| | - Morshed Kassouha
- Department of Microbiology, Faculty of Veterinary Medicine, Hama University, Hama, Syria
| | - Samer Ibrahim
- Department of Microbiology, Faculty of Veterinary Medicine, Hama University, Hama, Syria; Faculty of Dentistry, Arab Private University of science and Technology, Hama, Syria
| |
Collapse
|
9
|
Marchand T, Lamy T, Loughran TP. A modern view of LGL leukemia. Blood 2024; 144:1910-1923. [PMID: 38848524 DOI: 10.1182/blood.2023021790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/29/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
ABSTRACT Large granular lymphocytic leukemia (LGLL) is a rare lymphoproliferative chronic disorder characterized by expansion of either T or natural killer (NK) cytotoxic cells. In contrast to Epstein-Barr virus-induced aggressive NK-LGLL, chronic T-LGLL and NK-LGLL are indolent diseases affecting older patients with a median age of 66.5 years. LGLL is frequently associated with autoimmune disorders, most frequently rheumatoid arthritis. An auto-/alloantigen is tentatively implicated in disease initiation. Large granular lymphocyte expansion is then triggered by proinflammatory cytokines such as interleukin-15, macrophage inflammatory protein 1 (MIP-1), and RANTES (regulated upon activation, normal T cell expressed, and secreted). This proinflammatory environment contributes to deregulation of proliferative and apoptotic pathways. After the initial description of the JAK-STAT pathway signaling activation in the majority of patients, recurrent STAT3 gain-of-function mutations have been reported. The JAK-STAT pathway plays a key role in LGL pathogenesis by promoting survival, proliferation, and cytotoxicity. Several recent advances have been made toward understanding the molecular landscapes of T- and NK-LGLL, identifying multiple recurrent mutations affecting the epigenome, such as TET2 or KMT2D, and cross talk with the immune microenvironment, such as CCL22. Despite an indolent course, published series suggest that the majority of patients eventually need treatment. However, it is noteworthy that many patients may have a long-term observation period without ever requiring therapy. Treatments rely upon immunosuppressive drugs, namely cyclophosphamide, methotrexate, and cyclosporine. Recent advances have led to the development of targeted approaches, including JAK-STAT inhibitors, cytokine targeting, and hypomethylating agents, opening new developments in a still-incurable disease.
Collapse
Affiliation(s)
- Tony Marchand
- Department of Hematology, Rennes University Hospital, Rennes, France
- Faculty of Medicine, Rennes University, Rennes, France
- UMR 1236, Rennes University, INSERM, Établissement Français du Sang Bretagne, Rennes, France
| | - Thierry Lamy
- Department of Hematology, Rennes University Hospital, Rennes, France
- Faculty of Medicine, Rennes University, Rennes, France
- UMR 1236, Rennes University, INSERM, Établissement Français du Sang Bretagne, Rennes, France
| | - Thomas P Loughran
- Division of Hematology and Oncology, Department of Medicine and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
10
|
Gaur KK, Asuru TR, Srivastava M, Singh N, Purushotham N, Poojary B, Das B, Bhattacharyya S, Asthana S, Guchhait P. 7D, a small molecule inhibits dengue infection by increasing interferons and neutralizing-antibodies via CXCL4:CXCR3:p38:IRF3 and Sirt1:STAT3 axes respectively. EMBO Mol Med 2024; 16:2376-2401. [PMID: 39284947 PMCID: PMC11473809 DOI: 10.1038/s44321-024-00137-8] [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: 12/30/2023] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 10/16/2024] Open
Abstract
There are a limited number of effective vaccines against dengue virus (DENV) and significant efforts are being made to develop potent anti-virals. Previously, we described that platelet-chemokine CXCL4 negatively regulates interferon (IFN)-α/β synthesis and promotes DENV2 replication. An antagonist to CXCR3 (CXCL4 receptor) reversed it and inhibited viral replication. In a concurrent search, we identified CXCR3-antagonist from our compound library, namely 7D, which inhibited all serotypes of DENV in vitro. With a half-life of ~2.85 h in plasma and no significant toxicity, 7D supplementation (8 mg/kg-body-weight) to DENV2-infected IFNα/β/γR-/-AG129 or wild-type C57BL6 mice increased synthesis of IFN-α/β and IFN-λ, and rescued disease symptoms like thrombocytopenia, leukopenia and vascular-leakage, with improved survival. 7D, having the property to inhibit Sirt-1 deacetylase, promoted acetylation and phosphorylation of STAT3, which in-turn increased plasmablast proliferation, germinal-center maturation and synthesis of neutralizing-antibodies against DENV2 in mice. A STAT3-inhibitor successfully inhibited these effects of 7D. Together, these observations identify compound 7D as a stimulator of IFN-α/β/λ synthesis via CXCL4:CXCR3:p38:IRF3 signaling, and a booster for neutralizing-antibody generation by promoting STAT3-acetylation in plasmablasts, capable of protecting dengue infection.
Collapse
Affiliation(s)
- Kishan Kumar Gaur
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Tejeswara Rao Asuru
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Mitul Srivastava
- Translational Health Science Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Nitu Singh
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Nikil Purushotham
- Department of Studies in Chemistry, Mangalore University, Mangalagangotri, Karnataka, India
| | - Boja Poojary
- Department of Studies in Chemistry, Mangalore University, Mangalagangotri, Karnataka, India
| | - Bhabatosh Das
- Translational Health Science Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Sankar Bhattacharyya
- Translational Health Science Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Shailendra Asthana
- Translational Health Science Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India.
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India.
| |
Collapse
|
11
|
Rostami Nejad M, Razzaghi Z, Robati RM, Arjmand B, Rezaei-Tavirani M, Hamzeloo-Moghadam M, Keramatinia A. Molecular Mechanism Analysis of Intensive Light-Induced Retinal Damages. J Lasers Med Sci 2024; 15:e47. [PMID: 39450005 PMCID: PMC11499960 DOI: 10.34172/jlms.2024.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/07/2024] [Indexed: 10/26/2024]
Abstract
Introduction: The retina is a light-sensitive tissue, and intensive light exposure leads to light-induced retinal damage. It is pointed out that photoreceptor damage is responsible for the decrease in retina function. The aim of this study was to detect the main genes and biological terms which are involved in retinal response to intensive light exposure. Methods: The effect of intensive light on the mouse retina function was searched in the Gene Expression Omnibus (GEO) database. The data of GSE22818 were assessed by the GEO2R program. The significant differentially expressed genes (DEGs) were determined and evaluated via directed protein-protein interaction (PPI) network analysis. The critical significant DEGs were enriched via gene ontology analysis to find the related biological processes, molecular function, and biochemical pathways. Results: Data analysis indicates that the high intensity of light induces gene expression alteration in the retina. 105 significant DEGs were identified as the main responsive genes to light damage in the retina. STAT3, JUN, IL6ST, SOCS3, ATF3, JUNB, FOSL1, CCL2, ICAM1, FGF2, AGT, MYC, LIF, CISH, and EGR1 were introduced as the critical affected genes. STAT3, JUN, IL6ST, SOCS3, and ATF3 and "Positive regulation of the receptor signaling pathway via JAK-STAT" were highlighted as the key elements of molecular events. Conclusion: It can be concluded that regulation of the key DEGs and the dependent biological terms can effectively provide tools to prevent the development of light-induced retinal damage.
Collapse
Affiliation(s)
- Mohammad Rostami Nejad
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of medical sciences, Tehran, Iran
| | - Reza M Robati
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Iranian Cancer Control Center (MACSA), Tehran, Iran
| | | | - Maryam Hamzeloo-Moghadam
- Traditional Medicine and Materia Medica Research Center, School of Traditional Medicine Shahid, Beheshti University of Medical Sciences, Tehran, Iran
| | - Aliasghar Keramatinia
- Department of Community Medicine, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Gu X, Ju J, Chen Q, Ge M, Huang H. Investigation into the potential mechanism and therapeutic targets of Cangzhu Erchen decoction for the treatment of chronic obstructive pulmonary disease based on bioinformatics and network pharmacology. Medicine (Baltimore) 2024; 103:e39338. [PMID: 39151493 PMCID: PMC11332823 DOI: 10.1097/md.0000000000039338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 08/19/2024] Open
Abstract
This study aimed to elucidate the molecular mechanisms underlying the therapeutic effects of Cangzhu Erchen decoction (CZECD) in the treatment of chronic obstructive pulmonary disease (COPD) using microarray analysis, network pharmacology, and molecular docking. The active components and candidate targets of CZECD were obtained using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform and Swiss Target Prediction. COPD-related targets were collected from 5 databases. Access to drug-disease interface targets in the Venny platform. The Cytoscape program and the STRING database were used for protein-protein interaction analysis and subsequent core target screening. The DAVID database was used for Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes enrichment pathway analysis, while AutoDockTools was used for molecular docking to confirm binding affinity between drugs and key targets. A total of 140 compounds from CZECD and 5100 COPD-related targets were identified. SRC, PIK3CA, STAT3, PIK3R1, AKT1, HSP90AA1, PIK3CB, GRB2, PIK3CD, and MAPK1 were identified as the major targets of CZECD in its anti-COPD activity. GO and Kyoto Encyclopedia of Genes and Genomes enrichment studies revealed that CZECD mainly affects biological processes such as protein phosphorylation, xenobiotic response, positive regulation of the MAPK cascade, and inflammatory responses. Cancer, PI3K/AKT, and MAPK were the key pathways mediating these effects. The positive association between the core targets and the compounds was further validated by molecular docking. CZECD exerts its therapeutic role in COPD mainly through multiple compounds, targets, and pathways.
Collapse
Affiliation(s)
- Xiaofei Gu
- Department of Respiratory and Critical Care Medicine, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiangang Ju
- Department of Respiratory and Critical Care Medicine, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingqing Chen
- Department of Respiratory and Critical Care Medicine, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Minjie Ge
- Department of Respiratory and Critical Care Medicine, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huaqiong Huang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Ma CS, Tangye SG. STAT3 gain of function: Too much of a good thing in the skin! J Exp Med 2024; 221:e20240849. [PMID: 38949650 PMCID: PMC11215521 DOI: 10.1084/jem.20240849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Germline activating mutations in STAT3 cause a multi-systemic autoimmune and autoinflammatory condition. By studying a mouse model, Toth et al. (https://doi.org/10.1084/jem.20232091) propose a role for dysregulated IL-22 production by Th17 cells in causing some aspects of immune-mediated skin inflammation in human STAT3 GOF syndrome.
Collapse
Affiliation(s)
- Cindy S. Ma
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, UNSW Sydney, Darlinghurst, Australia
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, UNSW Sydney, Darlinghurst, Australia
| |
Collapse
|
14
|
Sobah ML, Liongue C, Ward AC. Stat3 Regulates Developmental Hematopoiesis and Impacts Myeloid Cell Function via Canonical and Non-Canonical Modalities. J Innate Immun 2024; 16:262-282. [PMID: 38643762 PMCID: PMC11249464 DOI: 10.1159/000538364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/12/2024] [Indexed: 04/23/2024] Open
Abstract
INTRODUCTION Signal transducer and activator of transcription (STAT) 3 is extensively involved in the development, homeostasis, and function of immune cells, with STAT3 disruption associated with human immune-related disorders. The roles ascribed to STAT3 have been assumed to be due to its canonical mode of action as an inducible transcription factor downstream of multiple cytokines, although alternative noncanonical functional modalities have also been identified. The relative involvement of each mode was further explored in relevant zebrafish models. METHODS Genome editing with CRISPR/Cas9 was used to generate mutants of the conserved zebrafish Stat3 protein: a loss of function knockout (KO) mutant and a mutant lacking C-terminal sequences including the transactivation domain (ΔTAD). Lines harboring these mutations were analyzed with respect to blood and immune cell development and function in comparison to wild-type zebrafish. RESULTS The Stat3 KO mutant showed perturbation of hematopoietic lineages throughout primitive and early definitive hematopoiesis. Neutrophil numbers did not increase in response to lipopolysaccharide (LPS) or granulocyte colony-stimulating factor (G-CSF) and their migration was significantly diminished, the latter correlating with abrogation of the Cxcl8b/Cxcr2 pathway, with macrophage responses perturbed. Intriguingly, many of these phenotypes were not shared by the Stat3 ΔTAD mutant. Indeed, only neutrophil and macrophage development were disrupted in these mutants with responsiveness to LPS and G-CSF maintained, and neutrophil migration actually increased. CONCLUSION This study has identified roles for zebrafish Stat3 within hematopoietic stem cells impacting multiple lineages throughout primitive and early definitive hematopoiesis, myeloid cell responses to G-CSF and LPS and neutrophil migration. Many of these roles showed conservation, but notably several involved noncanonical modalities, providing additional insights for relevant diseases.
Collapse
Affiliation(s)
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Alister C. Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| |
Collapse
|
15
|
Marchand T, Lamy T. The complex relationship between large granular lymphocyte leukemia and rheumatic disease. Expert Rev Clin Immunol 2024; 20:291-303. [PMID: 38105745 DOI: 10.1080/1744666x.2023.2292758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Large granular lymphocytic (LGL) leukemia is a rare lymphoproliferative disorder characterized by an expansion of clonal T or NK lymphocytes. Neutropenia-related infections represent the main clinical manifestation. Even if the disease follows an indolent course, most patients will ultimately need treatment in their lifetime. Interestingly, LGL leukemia is characterized by a high frequency of autoimmune disorders with rheumatoid arthritis being the most frequent. AREAS COVERED This review covers the pathophysiology, clinic-biological features and the advances made in the treatment of LGL leukemia. A special focus will be made on the similarities in the pathophysiology of LGL leukemia and the frequently associated rheumatic disorders. EXPERT OPINION Recent advances in the phenotypic and molecular characterization of LGL clones have uncovered the key role of JAK-STAT signaling in the pathophysiology linking leukemic cells expansion and autoimmunity. The description of the molecular landscape of T- and NK-LGL leukemia and the improved understanding of the associated rheumatic disorders open the way to the development of new targeted therapies effective on both conditions.
Collapse
Affiliation(s)
- Tony Marchand
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France
- Université Rennes 1, Rennes, France
- UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Thierry Lamy
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France
- Université Rennes 1, Rennes, France
- UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
- CIC 1414, Rennes, France
| |
Collapse
|
16
|
Tetarbe S, Shah K, Shah I. Intractable diarrhea in an infant-autoimmune enteropathy: A case report. JPGN REPORTS 2024; 5:70-73. [PMID: 38545278 PMCID: PMC10964334 DOI: 10.1002/jpr3.12038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 10/29/2023] [Accepted: 12/14/2023] [Indexed: 11/10/2024]
Abstract
Autoimmune enteropathy is a rare cause of chronic intractable diarrhea and is present in <1 in 100,000 infants. We report the case of a 9-month-old boy who presented with intractable diarrhea and vomiting. Genetic panel testing revealed a STAT3 heterozygous mutation in exon 6, suggesting infantile-onset multisystem autoimmune disease-1. The patient was initially treated with steroids and sulfasalazine. However, on tapering steroids, he had another episode of diarrhea and was subsequently put on baricitinib to which he responded.
Collapse
Affiliation(s)
- Shivangi Tetarbe
- Department of Pediatric Gastroenterology and HepatologyB J Wadia Hospital for ChildrenMumbaiIndia
| | | | - Ira Shah
- Department of Pediatric Gastroenterology and HepatologyB J Wadia Hospital for ChildrenMumbaiIndia
| |
Collapse
|
17
|
Liongue C, Sobah ML, Ward AC. Signal Transducer and Activator of Transcription Proteins at the Nexus of Immunodeficiency, Autoimmunity and Cancer. Biomedicines 2023; 12:45. [PMID: 38255152 PMCID: PMC10813391 DOI: 10.3390/biomedicines12010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
The signal transducer and activator of transcription (STAT) family of proteins has been demonstrated to perform pivotal roles downstream of a myriad of cytokines, particularly those that control immune cell production and function. This is highlighted by both gain-of-function (GOF) and loss-of-function (LOF) mutations being implicated in various diseases impacting cells of the immune system. These mutations are typically inherited, although somatic GOF mutations are commonly observed in certain immune cell malignancies. This review details the growing appreciation of STAT proteins as a key node linking immunodeficiency, autoimmunity and cancer.
Collapse
Affiliation(s)
- Clifford Liongue
- School of Medicine, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia; (C.L.); (M.L.S.)
- Institute for Mental and Physical Health and Clinical Translation, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia
| | - Mohamed Luban Sobah
- School of Medicine, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia; (C.L.); (M.L.S.)
| | - Alister C. Ward
- School of Medicine, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia; (C.L.); (M.L.S.)
- Institute for Mental and Physical Health and Clinical Translation, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia
| |
Collapse
|
18
|
Zerella JR, Homan CC, Arts P, Brown AL, Scott HS, Hahn CN. Transcription factor genetics and biology in predisposition to bone marrow failure and hematological malignancy. Front Oncol 2023; 13:1183318. [PMID: 37377909 PMCID: PMC10291195 DOI: 10.3389/fonc.2023.1183318] [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: 03/09/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Transcription factors (TFs) play a critical role as key mediators of a multitude of developmental pathways, with highly regulated and tightly organized networks crucial for determining both the timing and pattern of tissue development. TFs can act as master regulators of both primitive and definitive hematopoiesis, tightly controlling the behavior of hematopoietic stem and progenitor cells (HSPCs). These networks control the functional regulation of HSPCs including self-renewal, proliferation, and differentiation dynamics, which are essential to normal hematopoiesis. Defining the key players and dynamics of these hematopoietic transcriptional networks is essential to understanding both normal hematopoiesis and how genetic aberrations in TFs and their networks can predispose to hematopoietic disease including bone marrow failure (BMF) and hematological malignancy (HM). Despite their multifaceted and complex involvement in hematological development, advances in genetic screening along with elegant multi-omics and model system studies are shedding light on how hematopoietic TFs interact and network to achieve normal cell fates and their role in disease etiology. This review focuses on TFs which predispose to BMF and HM, identifies potential novel candidate predisposing TF genes, and examines putative biological mechanisms leading to these phenotypes. A better understanding of the genetics and molecular biology of hematopoietic TFs, as well as identifying novel genes and genetic variants predisposing to BMF and HM, will accelerate the development of preventative strategies, improve clinical management and counseling, and help define targeted treatments for these diseases.
Collapse
Affiliation(s)
- Jiarna R. Zerella
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Claire C. Homan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Peer Arts
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Anna L. Brown
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Hamish S. Scott
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Christopher N. Hahn
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
19
|
Ma CS, Tangye SG. Inborn errors of immunity: the Goldilocks effect-susceptibility to disease due to a little too much or a little too little. Clin Exp Immunol 2023; 212:93-95. [PMID: 37042727 PMCID: PMC10128157 DOI: 10.1093/cei/uxad039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 04/13/2023] Open
Affiliation(s)
- Cindy S Ma
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|