1
|
Buyukyilmaz G, Toksoy Adiguzel K, Kocaay P, Boyraz M, Metin A. Endocrinopathies in children with inborn errors of immunity: a single-center experience. J Pediatr Endocrinol Metab 2025:jpem-2024-0593. [PMID: 40270454 DOI: 10.1515/jpem-2024-0593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
OBJECTIVES Inborn errors of immunity (IEI) are a diverse group of genetically inherited disorders. We aimed to summarize and discuss endocrinopathies in children with IEI. METHODS This study included 84 IEI patients evaluated between September 2019 and September 2023. RESULTS We found that 15.6 % of the 32 patients with 22q11.2 deletion syndrome had permanent hypoparathyroidism. Hypergonadotropic hypogonadism was identified in one of four female patients with ataxia-telangiectasia (AT) and in all four females with severe congenital neutropenia (SCN) due to HAX1 deficiency. Additionally, hypergonadotropic hypogonadism was observed in one of nine males with common variable immunodeficiency (CVID). Among the CVID patients, one presented with autoimmune thyroiditis (AIT), type 1 diabetes mellitus (T1DM), hypoparathyroidism, and primary adrenal insufficiency. Of the 307 patients followed for selective IgA deficiency (sIgAD), 26 also received care in pediatric endocrinology. Among the sIgAD cases, 3.2 % had AIT and 4.5 % had T1DM. A patient with a STAT1 gain-of-function (GOF) variant was diagnosed with T1DM, AIT, and growth hormone deficiency, while a patient with a novel STAT3-GOF variant developed neonatal DM and interstitial lung disease. When the whole group was evaluated, thyroid disease was the most common endocrinopathy affecting 30.9 % of individuals, followed by DM, which was observed in 20.2 % of cases. CONCLUSIONS We have determined that AIT and T1DM were the most prevalent endocrine disorders in IEI patients. Pubertal development and gonadal functions should be monitored in the children with IEI. Early diagnosis and individualized treatment of endocrinopathies are crucial for a better quality of life and reduction of IEI-related complications.
Collapse
Affiliation(s)
- Gonul Buyukyilmaz
- Department of Pediatric Endocrinology, 639944 Ankara Bilkent City Hospital , Ankara, Türkiye
| | - Keziban Toksoy Adiguzel
- Department of Pediatric Endocrinology, 639944 Ankara Bilkent City Hospital , Ankara, Türkiye
- Department of Pediatric Endocrinology, Dr. Burhan Nalbantoğlu State Hospital, Nicosia, Northern CyprusTürkiye
| | - Pınar Kocaay
- Department of Pediatric Endocrinology, 639944 Ankara Bilkent City Hospital , Ankara, Türkiye
| | - Mehmet Boyraz
- Department of Pediatric Endocrinology, Ankara Yildirim Beyazit University Faculty of Medicine, Ankara, Türkiye
| | - Ayse Metin
- Department of Pediatric Allergy and Immunology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara, Türkiye
| |
Collapse
|
2
|
Hajialigol A, Azizi G, Seifi Alan M, Soltani A, Arabian Z, Tamiji M, Tsilifis C, Gennery AR, Jamee M. Dominant-Negative Versus Gain-of-Function STAT3 Defects: A Systematic Review on Epidemiological, Clinical, Immunological, and Molecular Aspects. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025:S2213-2198(25)00370-8. [PMID: 40246075 DOI: 10.1016/j.jaip.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Germline signal transducer and activator of transcription 3 (STAT3) mutations cause 2 distinct syndromes with predominant infectious or autoimmune phenotype. OBJECTIVE The objective of this study is to compile literature reports on gain-of-function (GOF) and dominant-negative (DN) mutations in the STAT3. METHODS We searched 3 main databases including PubMed, Scopus, and Web of Science from 1990 to 2023. All full-text articles and major reviews were manually searched for additional studies. RESULTS A total of 490 patients were reported in 107 articles including 265 patients with DN-STAT3 and 225 patients with GOF-STAT3 mutations. Major clinical differences between STAT3-DN and STAT3-GOF patients were observed in rates of infectious complication (98.2% vs 85.4%, P < .001), pneumonia (67.3% vs 52.1%, P = .006), sinusitis (17.5% vs 2.1%, P < .001), otitis (27.7% vs 5.2%, P < .001), abscess (52.7% vs 17.7%, P < .001), dermatologic manifestation (88.3% vs 58.4%, P < .001), atopic disorders (66.4% vs 40.2%, P < .001), interstitial lung disease (0.7% vs 17.7%, P < .001), dental/gingival disorders (38.5% vs 11.7%, P < .001), and endocrinopathies (0.9% vs 16.5%, P < .001), respectively. Most patients had normal counts of lymphocyte subsets, with the exceptions of higher CD3+ T cells and lower natural killer cells in STAT3-DN patients, and lower CD4+ T cells in STAT3-GOF patients. Most STAT3-DN patients had either normal or high serum concentrations of IgG, IgA, and IgM with almost universal raised serum IgE. Most STAT3-GOF patients had a low level of IgG, but normal levels of IgA, IgM, and IgE. CONCLUSIONS Patients with STAT3-DN had recurrent dermatological and pulmonary infections, eczema, elevated IgE, and eosinophilia, whereas patients with STAT3-GOF had early-onset polyautoimmunity and frequently require immunosuppressive therapy.
Collapse
Affiliation(s)
- Amirhossein Hajialigol
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Alborz Office of Universal Scientific Education and Research Network (USERN), Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Neurology, Thomas Jefferson University, Philadelphia, Pa
| | - Mahnaz Seifi Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Afsaneh Soltani
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Arabian
- School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mehrdad Tamiji
- Department of Computer Science, Rice University, Houston, Texas
| | - Christo Tsilifis
- Paediatric Immunology and Haematopoietic Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew R Gennery
- Paediatric Immunology and Haematopoietic Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Mahnaz Jamee
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
3
|
Meesilpavikkai K, Kaikaew K, Zhou Z, Dalm VA, Kaiser FM, Schliehe C, Swagemakers SM, van der Spek PJ, Schrijver B, Vasic P, de Bie M, Bakker M, Milanese C, Mastroberardino PG, Hirankarn N, Suratannon N, IJspeert H, Dik WA, Martin van Hagen P. Novel STAT3 Y360C Gain-of-function Variant Underlies Immune Dysregulation and Aberrancy in Mitochondrial Dynamics. Immune Netw 2025; 25:e18. [PMID: 40342844 PMCID: PMC12056293 DOI: 10.4110/in.2025.25.e18] [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: 10/27/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 05/11/2025] Open
Abstract
The STAT3 is an important regulator in a wide range of different cell types. Human STAT3 variants are associated with several immune dysregulation diseases. The current study investigated the clinical, genetic, and immunobiological data obtained from a family with novel heterozygous STAT3 variants located at p.Y360C of the DNA binding domain. The clinical manifestations of these patients include autoimmunity, immunodeficiency, and postnatal growth defects. Broad STAT3 regulated cells including patient primary immune cells and HEK293 cells harboring the variant were assessed. Remarkably high levels of STAT3-regulated cytokines were detected in the sera of the patients. STAT3 nuclear binding and STAT3 activity were higher in STAT3-transduced HEK293 cells containing the p.Y360C variant when compared to HEK cells expressing wild type (WT) STAT3. Upon cytokine activation, STAT3 variants inhibited nuclear translocation of the WT STAT3 molecule. We also demonstrated that PBMCs from these patients exhibit significantly higher mitochondrial activity compared to that of healthy controls. The exploration of the effects of STAT3 Y360C variants described in our study provides novel insights into the molecular effects of the STAT3 variant and its role in the pathophysiology of STAT3 gain-of-function syndromes.
Collapse
Affiliation(s)
- Kornvalee Meesilpavikkai
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Division of Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok10330, Thailand
| | - Kasiphak Kaikaew
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok10330, Thailand
| | - Zijun Zhou
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Division of Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Academic Center for Rare Immune Diseases (RIDC), Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Virgil A.S.H. Dalm
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Division of Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Academic Center for Rare Immune Diseases (RIDC), Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Fabian M.P. Kaiser
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Neonatal and Pediatric Intensive Care, Erasmus University Medical Center - Sophia Children’s Hospital, 3015 GD Rotterdam, The Netherlands
| | - Christopher Schliehe
- Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Sigrid M.A. Swagemakers
- Department of Pathology and Bioinformatics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Peter J. van der Spek
- Department of Pathology and Bioinformatics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Benjamin Schrijver
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Academic Center for Rare Immune Diseases (RIDC), Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Pamela Vasic
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Maaike de Bie
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Marleen Bakker
- Department of Pulmonary Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Chiara Milanese
- Department of Molecular Genetics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Pier G. Mastroberardino
- Department of Molecular Genetics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok10330, Thailand
| | - Narissara Suratannon
- Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok10330, Thailand
| | - Hanna IJspeert
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - P. Martin van Hagen
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Division of Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Academic Center for Rare Immune Diseases (RIDC), Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
4
|
Röring RJ, Scognamiglio F, de Jong LC, Groh LA, Matzaraki V, Koeken VACM, Joosten LAB, Ziogas A, Netea MG. Interleukin-10 inhibits important components of trained immunity in human monocytes. J Leukoc Biol 2025; 117:qiae240. [PMID: 39531639 DOI: 10.1093/jleuko/qiae240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/09/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Trained immunity induces antigen-agnostic enhancement of host defense and protection against secondary infections, but inappropriate activation can contribute to the pathophysiology of inflammatory diseases. Tight regulation of trained immunity is therefore needed to avoid pathology, but little is known about the endogenous processes that modulate it. Here, we investigated the potential of interleukin (IL)-10, a prototypical anti-inflammatory cytokine, to inhibit trained immunity. IL-10 induced tolerance and inhibited trained immunity in primary human monocytes at both functional and transcriptional levels. Inhibition of STAT3, a signaling route that mediates IL-10 signals, induced trained immunity. IL-10 downregulated glycolytic and oxidative metabolism in monocytes but did not impact the metabolic effects of β-glucan-induced trained immunity. Furthermore, IL-10 prevented increased reactive oxygen species production in Bacillus Calmette-Guérin (BCG)-induced training but did not influence phagocytosis upregulation. In a cohort study of healthy volunteers vaccinated with BCG, genetic variants that influenced IL-10 or its receptor modulated BCG-induced trained immunity. Furthermore, circulating IL-10 concentrations were negatively correlated with induction of trained immunity after BCG vaccination in a sex-specific manner. In conclusion, IL-10 inhibited several, albeit not all, immunological functions amplified after induction of trained immunity. Follow-up studies should explore the precise molecular mechanism that mediates the effects of IL-10 on trained immunity. Addressing these knowledge gaps is an important step toward optimizing IL-10's potential as a therapeutic target in diseases characterized by inappropriate induction of trained immunity.
Collapse
Affiliation(s)
- Rutger J Röring
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
- Inflammatory Origins Group, Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Road, 3052 Parkville, Australia
| | - Flavia Scognamiglio
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
| | - Lisanne C de Jong
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
| | - Laszlo A Groh
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
| | - Valerie A C M Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
- Research Centre Innovations in Care, Rotterdam University of Applied Sciences, Rochussenstraat 198, 3015 EK Rotterdam, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Strada Louis Pasteur 6, Cluj-Napoca 400535, Romania
| | - Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| |
Collapse
|
5
|
Dorando HK, Andrews JM, Borcherding NC, Quinn CC, Schmidt JA, Khatavkar OU, Aluri J, Harmon MT, Watkins MP, Frank A, Cooper MA, Musiek AC, Mehta-Shah N, Payton JE. Single Cell Resolution Tracking of Cutaneous T-Cell Lymphoma Reveals Clonal Evolution in Disease Progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637715. [PMID: 39990420 PMCID: PMC11844462 DOI: 10.1101/2025.02.11.637715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cutaneous T-cell lymphoma (CTCL) remains a challenging disease due to its significant heterogeneity, therapy resistance, and relentless progression. Multi-omics technologies offer the potential to provide uniquely precise views of disease progression and response to therapy. We present here a comprehensive multi-omics view of CTCL clonal evolution, incorporating exome, whole genome, epigenome, bulk-, single cell (sc) VDJ-, and scRNA-sequencing of 114 clinically annotated serial skin, peripheral blood, and lymph node samples from 35 CTCL patients. We leveraged this extensive dataset to define the molecular underpinnings of CTCL progression in individual patients at single cell resolution with the goal of identifying clinically useful biomarkers and therapeutic targets. Our studies identified a large number of recurrent progression-associated clonal genomic alterations; we highlight mutation of CCR4, PI3K signaling, and PD-1 checkpoint pathways as evasion tactics deployed by malignant T cells. We also identified a gain of function mutation in STAT3 (D661Y) and demonstrated by CUT&RUN-seq that it enhances binding to transcription start sites of genes in Rho GTPase pathways, which we previously reported to have activated chromatin and increased expression in HDACi-resistant CTCL. These data provide further support for a previously unrecognized role for Rho GTPase pathway dysregulation in CTCL progression. A striking number of progression-associated mutations occurred in chromatin methylation modifiers, including EZH2, suggesting that EZH1/2 inhibition may also benefit patients with CTCL. Knowledge of these molecular changes should be leveraged for improved disease monitoring, biomarker-informed clinical trial design, and new therapeutic strategies in this challenging and incurable cancer.
Collapse
|
6
|
Toth KA, Schmitt EG, Kolicheski A, Greenberg ZJ, Levendosky E, Saucier N, Trammel K, Oikonomou V, Lionakis MS, Klechevsky E, Kim BS, Schuettpelz LG, Saligrama N, Cooper MA. A human STAT3 gain-of-function variant drives local Th17 dysregulation and skin inflammation in mice. J Exp Med 2024; 221:e20232091. [PMID: 38861030 PMCID: PMC11167377 DOI: 10.1084/jem.20232091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/29/2024] [Accepted: 05/21/2024] [Indexed: 06/12/2024] Open
Abstract
Germline gain-of-function (GOF) variants in STAT3 cause an inborn error of immunity associated with early-onset poly-autoimmunity and immune dysregulation. To study tissue-specific immune dysregulation, we used a mouse model carrying a missense variant (p.G421R) that causes human disease. We observed spontaneous and imiquimod (IMQ)-induced skin inflammation associated with cell-intrinsic local Th17 responses in STAT3 GOF mice. CD4+ T cells were sufficient to drive skin inflammation and showed increased Il22 expression in expanded clones. Certain aspects of disease, including increased epidermal thickness, also required the presence of STAT3 GOF in epithelial cells. Treatment with a JAK inhibitor improved skin disease without affecting local Th17 recruitment and cytokine production. These findings collectively support the involvement of Th17 responses in the development of organ-specific immune dysregulation in STAT3 GOF and suggest that the presence of STAT3 GOF in tissues is important for disease and can be targeted with JAK inhibition.
Collapse
Affiliation(s)
- Kelsey A. Toth
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Erica G. Schmitt
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ana Kolicheski
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Zev J. Greenberg
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth Levendosky
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nermina Saucier
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kelsey Trammel
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vasileios Oikonomou
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Michail S. Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Eynav Klechevsky
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian S. Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, Precision Immunology Institute, Friedman Brain Institute, Mark Lebwohl Center for Neuroinflammation and Sensation, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, New York, NY, USA
| | - Laura G. Schuettpelz
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Naresha Saligrama
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- Bursky Center for Human Immunology & Immunotherapy, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan A. Cooper
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
7
|
Momenilandi M, Lévy R, Sobrino S, Li J, Lagresle-Peyrou C, Esmaeilzadeh H, Fayand A, Le Floc'h C, Guérin A, Della Mina E, Shearer D, Delmonte OM, Yatim A, Mulder K, Mancini M, Rinchai D, Denis A, Neehus AL, Balogh K, Brendle S, Rokni-Zadeh H, Changi-Ashtiani M, Seeleuthner Y, Deswarte C, Bessot B, Cremades C, Materna M, Cederholm A, Ogishi M, Philippot Q, Beganovic O, Ackermann M, Wuyts M, Khan T, Fouéré S, Herms F, Chanal J, Palterer B, Bruneau J, Molina TJ, Leclerc-Mercier S, Prétet JL, Youssefian L, Vahidnezhad H, Parvaneh N, Claeys KG, Schrijvers R, Luka M, Pérot P, Fourgeaud J, Nourrisson C, Poirier P, Jouanguy E, Boisson-Dupuis S, Bustamante J, Notarangelo LD, Christensen N, Landegren N, Abel L, Marr N, Six E, Langlais D, Waterboer T, Ginhoux F, Ma CS, Tangye SG, Meyts I, Lachmann N, Hu J, Shahrooei M, Bossuyt X, Casanova JL, Béziat V. FLT3L governs the development of partially overlapping hematopoietic lineages in humans and mice. Cell 2024; 187:2817-2837.e31. [PMID: 38701783 PMCID: PMC11149630 DOI: 10.1016/j.cell.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 03/04/2024] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
FMS-related tyrosine kinase 3 ligand (FLT3L), encoded by FLT3LG, is a hematopoietic factor essential for the development of natural killer (NK) cells, B cells, and dendritic cells (DCs) in mice. We describe three humans homozygous for a loss-of-function FLT3LG variant with a history of various recurrent infections, including severe cutaneous warts. The patients' bone marrow (BM) was hypoplastic, with low levels of hematopoietic progenitors, particularly myeloid and B cell precursors. Counts of B cells, monocytes, and DCs were low in the patients' blood, whereas the other blood subsets, including NK cells, were affected only moderately, if at all. The patients had normal counts of Langerhans cells (LCs) and dermal macrophages in the skin but lacked dermal DCs. Thus, FLT3L is required for B cell and DC development in mice and humans. However, unlike its murine counterpart, human FLT3L is required for the development of monocytes but not NK cells.
Collapse
Affiliation(s)
- Mana Momenilandi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Romain Lévy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Steicy Sobrino
- Laboratory of Chromatin and Gene Regulation During Development, Paris Cité University, UMR1163 INSERM, Imagine Institute, Paris, France; Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Jingwei Li
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Chantal Lagresle-Peyrou
- Paris Cité University, Imagine Institute, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Hossein Esmaeilzadeh
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Allergy and Clinical Immunology, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Antoine Fayand
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Sorbonne University, AP-HP, Tenon Hospital, Department of Internal Medicine, Paris, France
| | - Corentin Le Floc'h
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Antoine Guérin
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Erika Della Mina
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Debra Shearer
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France; Paris-Saclay University, Ile-de-France, France
| | - Mathieu Mancini
- Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Adeline Denis
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Karla Balogh
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Sarah Brendle
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Hassan Rokni-Zadeh
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran
| | - Majid Changi-Ashtiani
- School of Mathematics, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Caroline Deswarte
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Boris Bessot
- Paris Cité University, Imagine Institute, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Cassandre Cremades
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Omer Beganovic
- Laboratoire d'Onco-hématologie, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Mania Ackermann
- Hannover Medical School, Department of Pediatric Pulmonology, Allergology and Neonatology, Hannover, Germany; Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Margareta Wuyts
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
| | | | - Sébastien Fouéré
- Groupe Hospitalier Saint-Louis, Lariboisière, Fernand-Widal, CeGIDD, AP-HP, Paris, France
| | - Florian Herms
- Dermatology Department, Paris-Cité University, INSERM 976, Saint Louis Hospital, Paris, France
| | - Johan Chanal
- Dermatology Department, Cochin Hospital, INSERM U1016, AP-HP, Paris, France
| | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie Bruneau
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Thierry J Molina
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Stéphanie Leclerc-Mercier
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Jean-Luc Prétet
- Papillomavirus National Reference Center, Besançon Hospital, Besançon, France
| | - Leila Youssefian
- Department of Pathology and Laboratory Medicine, UCLA Clinical Genomics Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hassan Vahidnezhad
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nima Parvaneh
- Department of Pediatrics, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kristl G Claeys
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium; Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Rik Schrijvers
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Marine Luka
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Philippe Pérot
- Pathogen Discovery Laboratory, Institut Pasteur, Paris Cité University, Paris, France
| | - Jacques Fourgeaud
- Paris Cité University, URP 7328 FETUS, Paris, France; Microbiology Department, AP-HP, Necker Hospital for Sick Children, Paris, France
| | - Céline Nourrisson
- Clermont Auvergne University, INSERM U1071, M2iSH, USC INRAE 1382, CHU Clermont-Ferrand, 3IHP, Department of Parasitology-Mycology, Clermont-Ferrand, France; National Reference Center for Cryptosporidiosis, Microsporidia and Other Digestive Protozoa, Clermont-Ferrand, France
| | - Philippe Poirier
- Clermont Auvergne University, INSERM U1071, M2iSH, USC INRAE 1382, CHU Clermont-Ferrand, 3IHP, Department of Parasitology-Mycology, Clermont-Ferrand, France; National Reference Center for Cryptosporidiosis, Microsporidia and Other Digestive Protozoa, Clermont-Ferrand, France
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neil Christensen
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Centre for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Nico Marr
- Research Branch, Sidra Medicine, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Emmanuelle Six
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - David Langlais
- Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Tim Waterboer
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France; Paris-Saclay University, Ile-de-France, France
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Isabelle Meyts
- Laboratory of Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Department of Pediatrics, Leuven University Hospitals, Leuven, Belgium
| | - Nico Lachmann
- Hannover Medical School, Department of Pediatric Pulmonology, Allergology and Neonatology, Hannover, Germany; Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Jiafen Hu
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Mohammad Shahrooei
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium; Specialized Immunology Laboratory of Dr. Shahrooei, Tehran, Iran
| | - Xavier Bossuyt
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium; Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA; Howard Hughes Medical Institute, New York, NY, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.
| |
Collapse
|
8
|
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
|
9
|
Manoharan S, Saha S, Murugesan K, Santhakumar A, Perumal E. Natural bioactive compounds and STAT3 against hepatocellular carcinoma: An update. Life Sci 2024; 337:122351. [PMID: 38103726 DOI: 10.1016/j.lfs.2023.122351] [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] [Revised: 11/23/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is a challenging and very fatal liver cancer. The signal transducer and activator of transcription 3 (STAT3) pathway is a crucial regulator of tumor development and are ubiquitously active in HCC. Therefore, targeting STAT3 has emerged as a promising approach for preventing and treating HCC. Various natural bioactive compounds (NBCs) have been proven to target STAT3 and have the potential to prevent and treat HCC as STAT3 inhibitors. Numerous kinds of STAT3 inhibitors have been identified, including small molecule inhibitors, peptide inhibitors, and oligonucleotide inhibitors. Due to the undesirable side effects of the conventional therapeutic drugs against HCC, the focus is shifted to NBCs derived from plants and other natural sources. NBCs can be broadly classified into the categories of terpenes, alkaloids, carotenoids, and phenols. Most of the compounds belong to the family of terpenes, which prevent tumorigenesis by inhibiting STAT3 nuclear translocation. Further, through STAT3 inhibition, terpenes downregulate matrix metalloprotease 2 (MMP2), matrix metalloprotease 9 (MMP9) and vascular endothelial growth factor (VEGF), modulating metastasis. Terpenes also suppress the anti-apoptotic proteins and cell cycle markers. This review provides comprehensive information related to STAT3 abrogation by NBCs in HCC with in vitro and in vivo evidences.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Shreejit Saha
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Krishnasanthiya Murugesan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Aksayakeerthana Santhakumar
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India.
| |
Collapse
|
10
|
Arun S, Patel PK, Lakshmanan K, Rajangopal K, Swaminathan G, Byran G. Targeting STAT3 Enzyme for Cancer Treatment. Mini Rev Med Chem 2024; 24:1252-1261. [PMID: 38299278 DOI: 10.2174/0113895575254012231024062619] [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: 03/20/2023] [Revised: 08/03/2023] [Accepted: 09/14/2023] [Indexed: 02/02/2024]
Abstract
A category of cytoplasmic transcription factors called STATs mediates intracellular signaling, which is frequently generated at receptors on cell surfaces and subsequently sent to the nucleus. STAT3 is a member of a responsible for a variety of human tumor forms, including lymphomas, hematological malignancies, leukemias, multiple myeloma and several solid tumor types. Numerous investigations have demonstrated constitutive STAT3 activation lead to cancer development such as breast, head and neck, lung, colorectal, ovarian, gastric, hepatocellular, and prostate cancers. It's possible to get a hold of the book here. Tumor cells undergo apoptosis when STAT3 activation is suppressed. This review highlights the STAT3 activation and inhibition which can be used for further studies.
Collapse
Affiliation(s)
- Sowmiya Arun
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, 643001, Tamil Nadu, India
| | - Praveen Kumar Patel
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, 643001, Tamil Nadu, India
| | - Kaviarasan Lakshmanan
- Department of Pharmaceutical Chemistry, School of Pharmacy, Satyabhama Institute of Science and Technology, Chennai, India
| | - Kalirajan Rajangopal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, 643001, Tamil Nadu, India
| | - Gomathi Swaminathan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, 643001, Tamil Nadu, India
| | - Gowramma Byran
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, 643001, Tamil Nadu, India
| |
Collapse
|
11
|
Sobah ML, Liongue C, Ward AC. Contribution of Signal Transducer and Activator of Transcription 3 (STAT3) to Bone Development and Repair. Int J Mol Sci 2023; 25:389. [PMID: 38203559 PMCID: PMC10778865 DOI: 10.3390/ijms25010389] [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: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcription factor activated canonically by numerous cytokines and other factors, with significant roles in immunity, immune diseases, and cancer. It has also been implicated in several human skeletal disorders, with loss-of-function (LOF) mutations associated with aberrant skeletal development. To gain further insights, two zebrafish STAT3 lines were investigated: a complete LOF knockout (KO) mutant and a partial LOF mutant with the transactivation domain truncated (ΔTAD). Consistent with other studies, the KO mutants were smaller, with reduced length in early embryos exacerbated by a decreased growth rate from 5 days postfertilization (dpf). They displayed skeletal deformities that approached 80% incidence by 30 dpf, with a significant reduction in early bone but not cartilage formation. Further analysis additionally identified considerable abrogation of caudal fin regeneration, concomitant with a paucity of infiltrating macrophages and neutrophils, which may be responsible for this. Most of these phenotypes were also observed in the ΔTAD mutants, indicating that loss of canonical STAT3 signaling was the likely cause. However, the impacts on early bone formation and regeneration were muted in the ΔTAD mutant, suggesting the potential involvement of noncanonical functions in these processes.
Collapse
Affiliation(s)
- Mohamed L. Sobah
- School of Medicine, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| | - Clifford Liongue
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| | - Alister C. Ward
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| |
Collapse
|
12
|
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
|
13
|
Corbali O, Saxena S, Patel R, Lokhande H, Chitnis T. NF-κB and STAT3 activation in CD4 T cells in pediatric MOG antibody-associated disease. J Neuroimmunol 2023; 384:578197. [PMID: 37770354 DOI: 10.1016/j.jneuroim.2023.578197] [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: 03/03/2023] [Revised: 08/30/2023] [Accepted: 09/09/2023] [Indexed: 09/30/2023]
Abstract
In this study, we examined CD4 T cell activation using various stimuli in pediatric MOGAD patients (n = 4, untreated remission samples) and healthy controls (n = 5), to understand how both antigen-specific and bystander mechanisms contribute to CD4 T cell activation in MOGAD. TNFα, IL6, and MOG peptide pool were found to activate NF-κB or STAT3 pathways by measuring the expression of regulators (A20, IκBα) and phosphorylated subunits (phospho-p65 and phospho-STAT3) using immunolabeling. Prednisolone reversed activation of both NF-κB and STAT3 and increased the expression of A20 and IκBα. TNFR blocking partially reversed NF-κB activation in certain CD4 T cell subsets, but did not effect STAT3 activation. We observed that activation of NF-κB and STAT3 in response to various stimuli behaves mostly same in MOGAD (remission) and HC. IL6 stimulation resulted in higher STAT3 phosphorylation in MOGAD patients at 75 min, specifically in central and effector memory CD4 T cells (with unadjusted p-values). These findings suggest the potential therapeutic targeting of NF-κB and STAT3 pathways in MOGAD. Further investigation is needed to validate the significance of extended STAT3 phosphorylation and its correlation with IL6 receptor blocker treatment response.
Collapse
Affiliation(s)
- Osman Corbali
- Harvard Medical School, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Shrishti Saxena
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Rohit Patel
- Harvard Medical School, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Hrishikesh Lokhande
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tanuja Chitnis
- Harvard Medical School, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
14
|
Toskov V, Ehl S. Autoimmune lymphoproliferative immunodeficiencies (ALPID) in childhood: breakdown of immune homeostasis and immune dysregulation. Mol Cell Pediatr 2023; 10:11. [PMID: 37702894 PMCID: PMC10499775 DOI: 10.1186/s40348-023-00167-1] [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: 07/19/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
Many inborn errors of immunity (IEI) manifest with hallmarks of both immunodeficiency and immune dysregulation due to uncontrolled immune responses and impaired immune homeostasis. A subgroup of these disorders frequently presents with autoimmunity and lymphoproliferation (ALPID phenotype). After the initial description of the genetic basis of autoimmune lymphoproliferative syndrome (ALPS) more than 20 years ago, progress in genetics has helped to identify many more genetic conditions underlying this ALPID phenotype. Among these, the majority is caused by a group of autosomal-dominant conditions including CTLA-4 haploinsufficiency, STAT3 gain-of-function disease, activated PI3 kinase syndrome, and NF-κB1 haploinsufficiency. Even within a defined genetic condition, ALPID patients may present with staggering clinical heterogeneity, which makes diagnosis and management a challenge. In this review, we discuss the pathophysiology, clinical presentation, approaches to diagnosis, and conventional as well as targeted therapy of the most common ALPID conditions.
Collapse
Affiliation(s)
- Vasil Toskov
- Centre for Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stephan Ehl
- Centre for Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
15
|
Ashrafizadeh M, Mohan CD, Rangappa S, Zarrabi A, Hushmandi K, Kumar AP, Sethi G, Rangappa KS. Noncoding RNAs as regulators of STAT3 pathway in gastrointestinal cancers: Roles in cancer progression and therapeutic response. Med Res Rev 2023; 43:1263-1321. [PMID: 36951271 DOI: 10.1002/med.21950] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/09/2022] [Accepted: 02/28/2023] [Indexed: 03/24/2023]
Abstract
Gastrointestinal (GI) tumors (cancers of the esophagus, gastric, liver, pancreas, colon, and rectum) contribute to a large number of deaths worldwide. STAT3 is an oncogenic transcription factor that promotes the transcription of genes associated with proliferation, antiapoptosis, survival, and metastasis. STAT3 is overactivated in many human malignancies including GI tumors which accelerates tumor progression, metastasis, and drug resistance. Research in recent years demonstrated that noncoding RNAs (ncRNAs) play a major role in the regulation of many signaling pathways including the STAT3 pathway. The major types of endogenous ncRNAs that are being extensively studied in oncology are microRNAs, long noncoding RNAs, and circular RNAs. These ncRNAs can either be tumor-promoters or tumor-suppressors and each one of them imparts their activity via different mechanisms. The STAT3 pathway is also tightly modulated by ncRNAs. In this article, we have elaborated on the tumor-promoting role of STAT3 signaling in GI tumors. Subsequently, we have comprehensively discussed the oncogenic as well as tumor suppressor functions and mechanism of action of ncRNAs that are known to modulate STAT3 signaling in GI cancers.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chakrabhavi D Mohan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, India
| | - Shobith Rangappa
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri University, Nagamangala Taluk, India
| | - Ali Zarrabi
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Sariyer, Turkey
| | - Kiavash Hushmandi
- Division of Epidemiology, Faculty of Veterinary Medicine, Department of Food Hygiene and Quality Control, University of Tehran, Tehran, Iran
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
16
|
Abdullah SAA, Goa P, Vandenberghe E, Flavin R. Update on the Pathogenesis of Enteropathy-Associated T-Cell Lymphoma. Diagnostics (Basel) 2023; 13:2629. [PMID: 37627888 PMCID: PMC10453492 DOI: 10.3390/diagnostics13162629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
EATL is an aggressive T-cell non-Hodgkin lymphoma with poor prognosis and is largely localized to the small intestine. EATL is closely associated with coeliac disease (CD) and is seen mostly in patients originating from Northern Europe. Various factors are associated with an increased risk of developing EATL, such as viral infection, advanced age, being male, and the presence of the HLA-DQ2 haplotype. Clonal rearrangements in the TCR-β and γ genes have been reported in all EATL morphological variants with distinctive immunophenotypic characteristics. Although EATL can occur de novo, individuals with RCDII are at a higher risk of developing EATL. The cells of origin of EATL has been postulated to be normal small intestinal intraepithelial T-lymphocytes (IELs), and more recent evidence suggests a link between innate precursor IELs and EATL derived from refractory coeliac disease type II (RCDII). The immune microenvironment of mucosal cells within the small intestine enhances the process of neoplastic transformation of IELs into EATL. Cytokines such as IL-15 can activate and crucially deregulate the JAK-STAT signaling pathway by binding to receptors on the surface of IELs. Furthermore, mutations in the JAK/STAT pathway have been associated with RCDII-derived EATL.
Collapse
Affiliation(s)
| | - Patricia Goa
- Department of Histopathology, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
| | - Elisabeth Vandenberghe
- Department of Haematology, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
- Department of Haematology, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Richard Flavin
- Department of Histopathology, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Department of Histopathology, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
| |
Collapse
|
17
|
Schultz AB, Kugler DG, Nivelo L, Vitari N, Doyle LP, Ristin S, Hennighausen L, O’Shea JJ, Jankovic D, Villarino AV. T cell intrinsic STAT1 signaling prevents aberrant Th1 responses during acute toxoplasmosis. Front Immunol 2023; 14:1212190. [PMID: 37559725 PMCID: PMC10407301 DOI: 10.3389/fimmu.2023.1212190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/04/2023] [Indexed: 08/11/2023] Open
Abstract
Infection-induced T cell responses must be properly tempered and terminated to prevent immuno-pathology. Using transgenic mice, we demonstrate that T cell intrinsic STAT1 signaling is required to curb inflammation during acute infection with Toxoplasma gondii. Specifically, we report that mice lacking STAT1 selectively in T cells expel parasites but ultimately succumb to lethal immuno-pathology characterized by aberrant Th1-type responses with reduced IL-10 and increased IL-13 production. We also find that, unlike STAT1, STAT3 is not required for induction of IL-10 or suppression of IL-13 during acute toxoplasmosis. Each of these findings was confirmed in vitro and ChIP-seq data mining showed that STAT1 and STAT3 co-localize at the Il10 locus, as well as loci encoding other transcription factors that regulate IL-10 production, most notably Maf and Irf4. These data advance basic understanding of how infection-induced T cell responses are managed to prevent immuno-pathology and provide specific insights on the anti-inflammatory properties of STAT1, highlighting its role in shaping the character of Th1-type responses.
Collapse
Affiliation(s)
- Aaron B. Schultz
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
| | - David G. Kugler
- Immunoparasitology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Luis Nivelo
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
| | - Nicolas Vitari
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Laura P. Doyle
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Svetlana Ristin
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
| | - Lothar Hennighausen
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John J. O’Shea
- Lymphocyte Cell Biology Section, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dragana Jankovic
- Immunoparasitology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alejandro V. Villarino
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
| |
Collapse
|
18
|
Toth KA, Schmitt EG, Cooper MA. Deficiencies and Dysregulation of STAT Pathways That Drive Inborn Errors of Immunity: Lessons from Patients and Mouse Models of Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1463-1472. [PMID: 37126806 PMCID: PMC10151837 DOI: 10.4049/jimmunol.2200905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/11/2023] [Indexed: 05/03/2023]
Abstract
The STAT family proteins provide critical signals for immune cell development, differentiation, and proinflammatory and anti-inflammatory responses. Inborn errors of immunity (IEIs) are caused by single gene defects leading to immune deficiency and/or dysregulation, and they have provided opportunities to identify genes important for regulating the human immune response. Studies of patients with IEIs due to altered STAT signaling, and mouse models of these diseases, have helped to shape current understanding of the mechanisms whereby STAT signaling and protein interactions regulate immunity. Although many STAT signaling pathways are shared, clinical and immune phenotypes in patients with monogenic defects of STAT signaling highlight both redundant and nonredundant pathways. In this review, we provide an overview of the shared and unique signaling pathways used by STATs, phenotypes of IEIs with altered STAT signaling, and recent discoveries that have provided insight into the human immune response and treatment of disease.
Collapse
Affiliation(s)
- Kelsey A. Toth
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Erica G. Schmitt
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Megan A. Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
19
|
Ott N, Faletti L, Heeg M, Andreani V, Grimbacher B. JAKs and STATs from a Clinical Perspective: Loss-of-Function Mutations, Gain-of-Function Mutations, and Their Multidimensional Consequences. J Clin Immunol 2023:10.1007/s10875-023-01483-x. [PMID: 37140667 DOI: 10.1007/s10875-023-01483-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/01/2023] [Indexed: 05/05/2023]
Abstract
The JAK/STAT signaling pathway plays a key role in cytokine signaling and is involved in development, immunity, and tumorigenesis for nearly any cell. At first glance, the JAK/STAT signaling pathway appears to be straightforward. However, on closer examination, the factors influencing the JAK/STAT signaling activity, such as cytokine diversity, receptor profile, overlapping JAK and STAT specificity among non-redundant functions of the JAK/STAT complexes, positive regulators (e.g., cooperating transcription factors), and negative regulators (e.g., SOCS, PIAS, PTP), demonstrate the complexity of the pathway's architecture, which can be quickly disturbed by mutations. The JAK/STAT signaling pathway has been, and still is, subject of basic research and offers an enormous potential for the development of new methods of personalized medicine and thus the translation of basic molecular research into clinical practice beyond the use of JAK inhibitors. Gain-of-function and loss-of-function mutations in the three immunologically particularly relevant signal transducers STAT1, STAT3, and STAT6 as well as JAK1 and JAK3 present themselves through individual phenotypic clinical pictures. The established, traditional paradigm of loss-of-function mutations leading to immunodeficiency and gain-of-function mutation leading to autoimmunity breaks down and a more differentiated picture of disease patterns evolve. This review is intended to provide an overview of these specific syndromes from a clinical perspective and to summarize current findings on pathomechanism, symptoms, immunological features, and therapeutic options of STAT1, STAT3, STAT6, JAK1, and JAK3 loss-of-function and gain-of-function diseases.
Collapse
Affiliation(s)
- Nils Ott
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Laura Faletti
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Biological Sciences, Department of Molecular Biology, University of California, La Jolla, San Diego, CA, USA
| | - Virginia Andreani
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Clinic of Rheumatology and Clinical Immunology, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- DZIF - German Center for Infection Research, Satellite Center Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Freiburg, Germany
| |
Collapse
|
20
|
Begitt A, Krause S, Cavey JR, Vinkemeier DE, Vinkemeier U. A family-wide assessment of latent STAT transcription factor interactions reveals divergent dimer repertoires. J Biol Chem 2023; 299:104703. [PMID: 37059181 DOI: 10.1016/j.jbc.2023.104703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023] Open
Abstract
The conversion of STAT proteins from latent to active transcription factors is central to cytokine signalling. Triggered by their signal-induced tyrosine phosphorylation, it is the assembly of a range of cytokine-specific STAT homo- and heterodimers that marks a key step in the transition of hitherto latent proteins to transcription activators. In contrast, the constitutive self-assembly of latent STATs and how it relates to the functioning of activated STATs, is understood less well. To provide a more complete picture, we developed a co-localization-based assay and tested all 28 possible combinations of the seven unphosphorylated STAT (U-STAT) proteins in living cells. We identified five U-STAT homodimers -STAT1, STAT3, STAT4, STAT5A and STAT5B- and two heterodimers -STAT1:STAT2 and STAT5A:STAT5B- and performed semi-quantitative assessments of the forces and characterizations of binding interfaces that support them. One STAT protein -STAT6- was found to be monomeric. This comprehensive analysis of latent STAT self-assembly lays bare considerable structural and functional diversity in the ways that link STAT dimerization before and after activation.
Collapse
Affiliation(s)
- Andreas Begitt
- The University of Nottingham, School of Life Sciences, Nottingham, UK
| | - Sebastian Krause
- The University of Nottingham, School of Life Sciences, Nottingham, UK
| | - James R Cavey
- The University of Nottingham, School of Life Sciences, Nottingham, UK
| | | | - Uwe Vinkemeier
- The University of Nottingham, School of Life Sciences, Nottingham, UK
| |
Collapse
|
21
|
Peirolo A, Verolet C, Ranza E, Rohr M, Laurent M, Ruchonnet-Metrailler I, Worth AJJ, Blanchard-Rohner G. Hyper-IgE syndrome presenting with early life craniosynostosis in monozygotic twin sisters. Pediatr Allergy Immunol 2023; 34:e13944. [PMID: 37102391 DOI: 10.1111/pai.13944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 04/28/2023]
Affiliation(s)
- Anna Peirolo
- Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Charlotte Verolet
- Division of General Paediatrics, Department of Paediatrics, Gynaecology and Obstetrics, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Emmanuelle Ranza
- Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
| | - Marie Rohr
- Unit of Infectious Diseases, Division of General Paediatrics, Department of Paediatrics, Gynaecology and Obstetrics, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Meryle Laurent
- Unit of Paediatric Radiology, Department of Radiology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Isabelle Ruchonnet-Metrailler
- Unit of Paediatric Pneumology, Department of Paediatrics, Gynaecology and Obstetrics, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Austen J J Worth
- Department of Paediatric Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Geraldine Blanchard-Rohner
- Unit of Immunology and Vaccinology, Division of General Paediatrics, Department of Paediatrics, Gynaecology and Obstetrics, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| |
Collapse
|
22
|
Leiding JW, Vogel TP, Santarlas VGJ, Mhaskar R, Smith MR, Carisey A, Vargas-Hernández A, Silva-Carmona M, Heeg M, Rensing-Ehl A, Neven B, Hadjadj J, Hambleton S, Ronan Leahy T, Meesilpavikai K, Cunningham-Rundles C, Dutmer CM, Sharapova SO, Taskinen M, Chua I, Hague R, Klemann C, Kostyuchenko L, Morio T, Thatayatikom A, Ozen A, Scherbina A, Bauer CS, Flanagan SE, Gambineri E, Giovannini-Chami L, Heimall J, Sullivan KE, Allenspach E, Romberg N, Deane SG, Prince BT, Rose MJ, Bohnsack J, Mousallem T, Jesudas R, Santos Vilela MMD, O'Sullivan M, Pachlopnik Schmid J, Průhová Š, Klocperk A, Rees M, Su H, Bahna S, Baris S, Bartnikas LM, Chang Berger A, Briggs TA, Brothers S, Bundy V, Chan AY, Chandrakasan S, Christiansen M, Cole T, Cook MC, Desai MM, Fischer U, Fulcher DA, Gallo S, Gauthier A, Gennery AR, Gonçalo Marques J, Gottrand F, Grimbacher B, Grunebaum E, Haapaniemi E, Hämäläinen S, Heiskanen K, Heiskanen-Kosma T, Hoffman HM, Gonzalez-Granado LI, Guerrerio AL, Kainulainen L, Kumar A, Lawrence MG, Levin C, Martelius T, Neth O, Olbrich P, Palma A, Patel NC, Pozos T, Preece K, Lugo Reyes SO, Russell MA, Schejter Y, Seroogy C, Sinclair J, Skevofilax E, Suan D, Suez D, Szabolcs P, Velasco H, Warnatz K, Walkovich K, et alLeiding JW, Vogel TP, Santarlas VGJ, Mhaskar R, Smith MR, Carisey A, Vargas-Hernández A, Silva-Carmona M, Heeg M, Rensing-Ehl A, Neven B, Hadjadj J, Hambleton S, Ronan Leahy T, Meesilpavikai K, Cunningham-Rundles C, Dutmer CM, Sharapova SO, Taskinen M, Chua I, Hague R, Klemann C, Kostyuchenko L, Morio T, Thatayatikom A, Ozen A, Scherbina A, Bauer CS, Flanagan SE, Gambineri E, Giovannini-Chami L, Heimall J, Sullivan KE, Allenspach E, Romberg N, Deane SG, Prince BT, Rose MJ, Bohnsack J, Mousallem T, Jesudas R, Santos Vilela MMD, O'Sullivan M, Pachlopnik Schmid J, Průhová Š, Klocperk A, Rees M, Su H, Bahna S, Baris S, Bartnikas LM, Chang Berger A, Briggs TA, Brothers S, Bundy V, Chan AY, Chandrakasan S, Christiansen M, Cole T, Cook MC, Desai MM, Fischer U, Fulcher DA, Gallo S, Gauthier A, Gennery AR, Gonçalo Marques J, Gottrand F, Grimbacher B, Grunebaum E, Haapaniemi E, Hämäläinen S, Heiskanen K, Heiskanen-Kosma T, Hoffman HM, Gonzalez-Granado LI, Guerrerio AL, Kainulainen L, Kumar A, Lawrence MG, Levin C, Martelius T, Neth O, Olbrich P, Palma A, Patel NC, Pozos T, Preece K, Lugo Reyes SO, Russell MA, Schejter Y, Seroogy C, Sinclair J, Skevofilax E, Suan D, Suez D, Szabolcs P, Velasco H, Warnatz K, Walkovich K, Worth A, Seppänen MRJ, Torgerson TR, Sogkas G, Ehl S, Tangye SG, Cooper MA, Milner JD, Forbes Satter LR. Monogenic early-onset lymphoproliferation and autoimmunity: Natural history of STAT3 gain-of-function syndrome. J Allergy Clin Immunol 2023; 151:1081-1095. [PMID: 36228738 PMCID: PMC10081938 DOI: 10.1016/j.jaci.2022.09.002] [Show More Authors] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND In 2014, germline signal transducer and activator of transcription (STAT) 3 gain-of-function (GOF) mutations were first described to cause a novel multisystem disease of early-onset lymphoproliferation and autoimmunity. OBJECTIVE This pivotal cohort study defines the scope, natural history, treatment, and overall survival of a large global cohort of patients with pathogenic STAT3 GOF variants. METHODS We identified 191 patients from 33 countries with 72 unique mutations. Inclusion criteria included symptoms of immune dysregulation and a biochemically confirmed germline heterozygous GOF variant in STAT3. RESULTS Overall survival was 88%, median age at onset of symptoms was 2.3 years, and median age at diagnosis was 12 years. Immune dysregulatory features were present in all patients: lymphoproliferation was the most common manifestation (73%); increased frequencies of double-negative (CD4-CD8-) T cells were found in 83% of patients tested. Autoimmune cytopenias were the second most common clinical manifestation (67%), followed by growth delay, enteropathy, skin disease, pulmonary disease, endocrinopathy, arthritis, autoimmune hepatitis, neurologic disease, vasculopathy, renal disease, and malignancy. Infections were reported in 72% of the cohort. A cellular and humoral immunodeficiency was observed in 37% and 51% of patients, respectively. Clinical symptoms dramatically improved in patients treated with JAK inhibitors, while a variety of other immunomodulatory treatment modalities were less efficacious. Thus far, 23 patients have undergone bone marrow transplantation, with a 62% survival rate. CONCLUSION STAT3 GOF patients present with a wide array of immune-mediated disease including lymphoproliferation, autoimmune cytopenias, and multisystem autoimmunity. Patient care tends to be siloed, without a clear treatment strategy. Thus, early identification and prompt treatment implementation are lifesaving for STAT3 GOF syndrome.
Collapse
Affiliation(s)
- Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore; Johns Hopkins All Children's Institute for Clinical and Translational Research, Johns Hopkins All Children's Hospital, St Petersburg.
| | - Tiphanie P Vogel
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston
| | | | - Rahul Mhaskar
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa
| | - Madison R Smith
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston
| | - Alexandre Carisey
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis
| | - Alexander Vargas-Hernández
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston
| | - Manuel Silva-Carmona
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Anne Rensing-Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Bénédicte Neven
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163-Institut Imagine, Paris
| | - Jérôme Hadjadj
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163-Institut Imagine, Paris
| | - Sophie Hambleton
- Newcastle University Translational and Clinical Research Institute, Newcastle (United Kingdom)
| | | | - Kornvalee Meesilpavikai
- Department of Internal Medicine, Division of Clinical Immunology and Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Cullen M Dutmer
- Children's Hospital Colorado, University of Colorado School of Medicine, Aurora
| | - Svetlana O Sharapova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk
| | - Mervi Taskinen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Turku and Kuopio, Finland
| | - Ignatius Chua
- Department of Rheumatology, Immunology and Allergy, Christchurch Hospital, Christchurch; Clinical Immunogenomics Research Consortium of Australasia (CIRCA)
| | | | - Christian Klemann
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover
| | - Larysa Kostyuchenko
- Center of Pediatric Immunology, Western Ukrainian Specialized Children's Medical Centre, Lviv
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo
| | - Akaluck Thatayatikom
- Division of Pediatric Allergy/Immunology/Rheumatology, Shands Children's Hospital, University of Florida, Gainesville
| | - Ahmet Ozen
- School of Medicine, Pediatric Allergy and Immunology, Marmara University, Istanbul
| | - Anna Scherbina
- Dmitry Rogachev National Medical and Research Center for Pediatric Hematology, Oncology and Immunology, Moscow
| | - Cindy S Bauer
- Division of Allergy and Immunology, Phoenix Children's Hospital, Phoenix
| | - Sarah E Flanagan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter
| | - Eleonora Gambineri
- Department of NEUROFARBA, Section of Children's Health, University of Florence, Anna Meyer Children's Hospital, Florence
| | | | - Jennifer Heimall
- Perelman School of Medicine at University of Pennsylvania, Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia
| | - Kathleen E Sullivan
- Perelman School of Medicine at University of Pennsylvania, Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia
| | - Eric Allenspach
- Pediatric Immunology/Rheumatology, University of Washington, Seattle; Seattle Children's Hospital, Seattle
| | - Neil Romberg
- Perelman School of Medicine at University of Pennsylvania, Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia
| | - Sean G Deane
- Department of Allergy, The Permanente Medical Group, Sacramento, and the Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, School of Medicine, Sacramento
| | - Benjamin T Prince
- Nationwide Children's Hospital Department of Allergy and Immunology, Columbus; College of Medicine, The Ohio State University, Columbus
| | - Melissa J Rose
- College of Medicine, The Ohio State University, Columbus; Division of Pediatric Hematology-Oncology, Nationwide Children's Hospital, Columbus
| | - John Bohnsack
- Department of Pediatrics, University of Utah, Salt Lake City
| | | | - Rohith Jesudas
- Department of Hematology, St Jude Children's Research Hospital, Memphis
| | - Maria Marluce Dos Santos Vilela
- Pediatric Allergy and Immunology/Center of Investigation in Pediatrics, Faculty of Medical Sciences, State University of Campinas-Unicamp, São Paulo
| | - Michael O'Sullivan
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Immunology Department, Perth Children's Hospital, Nedlands
| | - Jana Pachlopnik Schmid
- Division of Immunology, University Children's Hospital Zurich, Children's Research Center (CRC), Zurich
| | - Štěpánka Průhová
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, Prague
| | - Adam Klocperk
- Department of Immunology, Second Faculty of Medicine and University Hospital Motol, Charles University in Prague, Prague
| | - Matthew Rees
- Department of Hematology, St Jude Children's Research Hospital, Memphis
| | - Helen Su
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda
| | - Sami Bahna
- Allergy and Immunology Section, Louisiana State University Health Sciences Center, Shreveport
| | - Safa Baris
- School of Medicine, Pediatric Allergy and Immunology, Marmara University, Istanbul
| | - Lisa M Bartnikas
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston
| | - Amy Chang Berger
- Division of Hospital Medicine, Department of Medicine, University of California, San Francisco
| | - Tracy A Briggs
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester; NW Genomic Laboratory Hub, Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester
| | - Shannon Brothers
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Starship Children's Hospital, Auckland
| | - Vanessa Bundy
- Allergy and Immunology, University of California, Los Angeles
| | - Alice Y Chan
- Department of Medicine, University of California, San Francisco
| | - Shanmuganathan Chandrakasan
- Division of Bone Marrow Transplant, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta
| | | | - Theresa Cole
- Department of Allergy and Immunology, The Royal Children's Hospital, Melbourne
| | - Matthew C Cook
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra
| | | | - Ute Fischer
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf
| | - David A Fulcher
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra
| | - Silvanna Gallo
- Department of Pediatrics, Immunology and Rheumatology Section, Puerto Montt Hospital, Puerto Montt
| | - Amelie Gauthier
- Department of Allergy and Immunology, CHU de Québec-CHUL, Laval University Hospital Center, Laval University, Quebec City
| | - Andrew R Gennery
- Newcastle University Translational and Clinical Research Institute, Newcastle (United Kingdom)
| | - José Gonçalo Marques
- Infectious Diseases and Immunodeficiencies Unit, Department of Pediatrics, Hospital de Santa Maria-CHULN and Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Frédéric Gottrand
- University Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, Lille
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Eyal Grunebaum
- Division of Immunology and Allergy, and the Department of Pediatrics, Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto
| | - Emma Haapaniemi
- Centre for Molecular Medicine Norway, Oslo; Department of Pediatric Research, Oslo
| | | | - Kaarina Heiskanen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Turku and Kuopio, Finland
| | | | - Hal M Hoffman
- Department of Pediatrics, University of California San Diego, La Jolla; Rady Children's Hospital San Diego, Division of Pediatric Allergy, Immunology, and Rheumatology, San Diego
| | - Luis Ignacio Gonzalez-Granado
- Pediatrics Department, University Hospital 12 de Octubre, Research Institute Hospital, School of Medicine Complutense University, Madrid
| | - Anthony L Guerrerio
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore
| | - Leena Kainulainen
- Department of Pediatrics and Medicine, Turku University Hospital, University of Turku, Turku, Finland
| | - Ashish Kumar
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati
| | | | - Carina Levin
- Pediatric Hematology Unit, Emek Medical Centre, Afula, and the Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa
| | - Timi Martelius
- Adult Immunodeficiency Unit, Inflammation Center, Helsinki University Hospital and University of Helsinki, Helsinki
| | - Olaf Neth
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Peter Olbrich
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Alejandro Palma
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof Dr Juan P. Garrahan, Buenos Aires
| | - Niraj C Patel
- Division of Allergy and Immunology, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta
| | - Tamara Pozos
- Department of Clinical Immunology, Children's Minnesota, Minneapolis
| | - Kahn Preece
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Department of Paediatric Immunology, John Hunter Children's Hospital, Newcastle (Australia)
| | | | | | - Yael Schejter
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Ein-Kerem Medical Center and Faculty of Medicine, Hebrew University, Jerusalem
| | - Christine Seroogy
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison
| | - Jan Sinclair
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Starship Children's Hospital, Auckland
| | - Effie Skevofilax
- Department of Pediatric Hematology-Oncology (TAO) and First Department of Pediatrics, Aghia Sophia Children's Hospital, Athens
| | - Daniel Suan
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Garvan Institute of Medical Research, Darlinghurst; Westmead Clinical School, University of Sydney, Westmead
| | - Daniel Suez
- Allergy, Asthma & Immunology Clinic, PA, Irving
| | - Paul Szabolcs
- University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, Pittsburgh
| | - Helena Velasco
- Division of Allergy and Clinical Immunology, Moinhos de Vento Hospital, Porto Alegre
| | - Klaus Warnatz
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Kelly Walkovich
- Department of Pediatrics, C. S. Mott Children's Hospital, Michigan Medicine, Ann Arbor
| | - Austen Worth
- Great Ormond Street Hospital for Children, London
| | - Mikko R J Seppänen
- Rare Disease Center, Children's Hospital, and Adult Primary Immunodeficiency Outpatient Clinic, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki
| | | | - Georgios Sogkas
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hanover
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Stuart G Tangye
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Garvan Institute of Medical Research, Darlinghurst; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology and Immunology, Washington University School of Medicine, St Louis
| | - Joshua D Milner
- Department of Pediatrics, Division of Allergy and Immunology, Columbia University, New York Presbyterian Hospital, New York
| | - Lisa R Forbes Satter
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston.
| |
Collapse
|
23
|
Heidary S, Awasthi N, Page N, Allnutt T, Lewis RS, Liongue C, Ward AC. A zebrafish model of growth hormone insensitivity syndrome with immune dysregulation 1 (GHISID1). Cell Mol Life Sci 2023; 80:109. [PMID: 36995466 PMCID: PMC10063521 DOI: 10.1007/s00018-023-04759-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/03/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
Signal transducer and activator of transcription (STAT) proteins act downstream of cytokine receptors to facilitate changes in gene expression that impact a range of developmental and homeostatic processes. Patients harbouring loss-of-function (LOF) STAT5B mutations exhibit postnatal growth failure due to lack of responsiveness to growth hormone as well as immune perturbation, a disorder called growth hormone insensitivity syndrome with immune dysregulation 1 (GHISID1). This study aimed to generate a zebrafish model of this disease by targeting the stat5.1 gene using CRISPR/Cas9 and characterising the effects on growth and immunity. The zebrafish Stat5.1 mutants were smaller, but exhibited increased adiposity, with concomitant dysregulation of growth and lipid metabolism genes. The mutants also displayed impaired lymphopoiesis with reduced T cells throughout the lifespan, along with broader disruption of the lymphoid compartment in adulthood, including evidence of T cell activation. Collectively, these findings confirm that zebrafish Stat5.1 mutants mimic the clinical impacts of human STAT5B LOF mutations, establishing them as a model of GHISID1.
Collapse
Affiliation(s)
- Somayyeh Heidary
- School of Medicine, Deakin University, Pigdons Road, Geelong, VIC, 3216, Australia
| | - Nagendra Awasthi
- School of Medicine, Deakin University, Pigdons Road, Geelong, VIC, 3216, Australia
| | - Nicole Page
- School of Medicine, Deakin University, Pigdons Road, Geelong, VIC, 3216, Australia
| | - Theo Allnutt
- School of Medicine, Deakin University, Pigdons Road, Geelong, VIC, 3216, Australia
| | - Rowena S Lewis
- School of Life and Environmental Sciences, Deakin University, Burwood, VIC, 3125, Australia
| | - Clifford Liongue
- School of Medicine, Deakin University, Pigdons Road, Geelong, VIC, 3216, Australia
- IMPACT, Deakin University, Geelong, VIC, 3216, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Pigdons Road, Geelong, VIC, 3216, Australia.
- IMPACT, Deakin University, Geelong, VIC, 3216, Australia.
| |
Collapse
|
24
|
Bogaert DJ, Kuehn HS, Bordon V, Haerynck F. Editorial: The role of transcription factors in inborn errors of immunity. Front Immunol 2023; 14:1189312. [PMID: 37051249 PMCID: PMC10083482 DOI: 10.3389/fimmu.2023.1189312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Affiliation(s)
- Delfien J. Bogaert
- Department of Pediatrics, Division of Pediatric Hemato-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Primary Immunodeficiency (PID) Research Laboratory, Center for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Center, Ghent University, Ghent, Belgium
| | - Hye Sun Kuehn
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health, Clinical Center, Bethesda, MD, United States
| | - Victoria Bordon
- Department of Pediatrics, Division of Pediatric Hemato-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, Primary Immunodeficiency (PID) Research Laboratory, Center for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Center, Ghent University, Ghent, Belgium
- Department of Pediatrics, Division of Pediatric Pulmonology, Immunology and Infectious Diseases, Ghent University Hospital, Ghent, Belgium
- *Correspondence: Filomeen Haerynck,
| |
Collapse
|
25
|
Mansouri M, El Haddoumi G, Bendani H, Boumajdi N, Hakmi M, Abbou H, Bouricha EM, Elgharbaoui B, Kartti S, El Jaoudi R, Belyamani L, Kandoussi I, Ibrahimi A, El Hafidi N. In Silico Analyses of All STAT3 Missense Variants Leading to Explore Divergent AD-HIES Clinical Phenotypes. Evol Bioinform Online 2023; 19:11769343231169374. [PMID: 37123531 PMCID: PMC10134169 DOI: 10.1177/11769343231169374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/20/2023] [Indexed: 05/02/2023] Open
Abstract
Autosomal dominant hyper-IgE syndrome (AD-HIES) is linked to dominant negative mutations of the STAT3 protein whose molecular basis for dysfunction is unclear and presenting with a variety of clinical manifestations with only supportive treatment. To establish the relationship between the impact of STAT3 mutations in different domains and the severity of the clinical manifestations, 105 STAT3 mutations were analyzed for their impact on protein stability, flexibility, function, and binding affinity using in Silico approaches. Our results showed that 73% of the studied mutations have an impact on the physicochemical properties of the protein, altering the stability, flexibility and function to varying degrees. In particular, mutations affecting the DNA binding domain (DBD) and the Src Homology 2 (SH2) have a significant impact on the protein structure and disrupt its interaction either with DNA or other STAT3 to form a heterodomain complex, leading to severe clinical phenotypes. Collectively, this study suggests that there is a close relationship between the domain involving the mutation, the degree of variation in the properties of the protein and the degree of loss of function ranging from partial loss to complete loss, explaining the variability of clinical manifestations between mild and severe.
Collapse
Affiliation(s)
- Mariam Mansouri
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Ghyzlane El Haddoumi
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Houda Bendani
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Nasma Boumajdi
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Mohammed Hakmi
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Hanane Abbou
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
- Mohammed VI University of Health
Sciences (UM6SS), Casablanca, Morocco
| | - El Mehdi Bouricha
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Boutaina Elgharbaoui
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Souad Kartti
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Rachid El Jaoudi
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
- Pharmacology and Toxicology Department,
Faculty of Medicine and Pharmacy, University Mohamed V, Rabat, Morocco
| | - Lahcen Belyamani
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
- Mohammed VI University of Health
Sciences (UM6SS), Casablanca, Morocco
- Emergency Department, Military Hospital
Mohammed V, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Ilham Kandoussi
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
| | - Azeddine Ibrahimi
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
- Mohammed VI University of Health
Sciences (UM6SS), Casablanca, Morocco
| | - Naima El Hafidi
- Biotechnology lab (MedBiotech),
Bioinova Research Center, Medical and Pharmacy School, Mohammed V University in
Rabat, Morocco
- Centre Mohammed VI of Research and
Innovation (CM6), Rabat, Morocco
- Division of Pediatric immunoallergology
and Infectious Diseases, Children University Hospital, Ibn Sina University Hospital,
Rabat, Morocco
- Pr. Naima El Hafidi, Biotechnology lab
(MedBiotech), Bioinova Research Center, Medical & Pharmacy School, Mohammed
V university in Rabat, Imp. Souissi, Rabat 10100, Morocco.
| |
Collapse
|
26
|
Çetin G, Studencka-Turski M, Venz S, Schormann E, Junker H, Hammer E, Völker U, Ebstein F, Krüger E. Immunoproteasomes control activation of innate immune signaling and microglial function. Front Immunol 2022; 13:982786. [PMID: 36275769 PMCID: PMC9584546 DOI: 10.3389/fimmu.2022.982786] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and play a major role in the regulation of brain homeostasis. To maintain their cellular protein homeostasis, microglia express standard proteasomes and immunoproteasomes (IP), a proteasome isoform that preserves protein homeostasis also in non-immune cells under challenging conditions. The impact of IP on microglia function in innate immunity of the CNS is however not well described. Here, we establish that IP impairment leads to proteotoxic stress and triggers the unfolded and integrated stress responses in mouse and human microglia models. Using proteomic analysis, we demonstrate that IP deficiency in microglia results in profound alterations of the ubiquitin-modified proteome among which proteins involved in the regulation of stress and immune responses. In line with this, molecular analysis revealed chronic activation of NF-κB signaling in IP-deficient microglia without further stimulus. In addition, we show that IP impairment alters microglial function based on markers for phagocytosis and motility. At the molecular level IP impairment activates interferon signaling promoted by the activation of the cytosolic stress response protein kinase R. The presented data highlight the importance of IP function for the proteostatic potential as well as for precision proteolysis to control stress and immune signaling in microglia function.
Collapse
Affiliation(s)
- Gonca Çetin
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Maja Studencka-Turski
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Simone Venz
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Eileen Schormann
- Institute of Biochemistry, Charité – University Medicine Berlin, Berlin, Germany
| | - Heike Junker
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Frédéric Ebstein
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
- *Correspondence: Elke Krüger,
| |
Collapse
|
27
|
Campos JS, Henrickson SE. Defining and targeting patterns of T cell dysfunction in inborn errors of immunity. Front Immunol 2022; 13:932715. [PMID: 36189259 PMCID: PMC9516113 DOI: 10.3389/fimmu.2022.932715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Inborn errors of immunity (IEIs) are a group of more than 450 monogenic disorders that impair immune development and function. A subset of IEIs blend increased susceptibility to infection, autoimmunity, and malignancy and are known collectively as primary immune regulatory disorders (PIRDs). While many aspects of immune function are altered in PIRDs, one key impact is on T-cell function. By their nature, PIRDs provide unique insights into human T-cell signaling; alterations in individual signaling molecules tune downstream signaling pathways and effector function. Quantifying T-cell dysfunction in PIRDs and the underlying causative mechanisms is critical to identifying existing therapies and potential novel therapeutic targets to treat our rare patients and gain deeper insight into the basic mechanisms of T-cell function. Though there are many types of T-cell dysfunction, here we will focus on T-cell exhaustion, a key pathophysiological state. Exhaustion has been described in both human and mouse models of disease, where the chronic presence of antigen and inflammation (e.g., chronic infection or malignancy) induces a state of altered immune profile, transcriptional and epigenetic states, as well as impaired T-cell function. Since a subset of PIRDs amplify T-cell receptor (TCR) signaling and/or inflammatory cytokine signaling cascades, it is possible that they could induce T-cell exhaustion by genetically mimicking chronic infection. Here, we review the fundamentals of T-cell exhaustion and its possible role in IEIs in which genetic mutations mimic prolonged or amplified T-cell receptor and/or cytokine signaling. Given the potential insight from the many forms of PIRDs in understanding T-cell function and the challenges in obtaining primary cells from these rare disorders, we also discuss advances in CRISPR-Cas9 genome-editing technologies and potential applications to edit healthy donor T cells that could facilitate further study of mechanisms of immune dysfunctions in PIRDs. Editing T cells to match PIRD patient genetic variants will allow investigations into the mechanisms underpinning states of dysregulated T-cell function, including T-cell exhaustion.
Collapse
Affiliation(s)
- Jose S. Campos
- Division of Allergy and Immunology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Sarah E. Henrickson
- Division of Allergy and Immunology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
28
|
Guevara-Hoyer K, Fuentes-Antrás J, de la Fuente-Muñoz E, Fernández-Arquero M, Solano F, Pérez-Segura P, Neves E, Ocaña A, Pérez de Diego R, Sánchez-Ramón S. Genomic crossroads between non-Hodgkin's lymphoma and common variable immunodeficiency. Front Immunol 2022; 13:937872. [PMID: 35990641 PMCID: PMC9390007 DOI: 10.3389/fimmu.2022.937872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/07/2022] [Indexed: 12/03/2022] Open
Abstract
Common variable immunodeficiency (CVID) represents the largest group of primary immunodeficiencies that may manifest with infections, inflammation, autoimmunity, and cancer, mainly B-cell non-Hodgkin's lymphoma (NHL). Indeed, NHL may result from chronic or recurrent infections and has, therefore, been recognized as a clinical phenotype of CVID, although rare. The more one delves into the mechanisms involved in CVID and cancer, the stronger the idea that both pathologies can be a reflection of the same primer events observed from different angles. The potential effects of germline variants on specific somatic modifications in malignancies suggest that it might be possible to anticipate critical events during tumor development. In the same way, a somatic alteration in NHL could be conditioning a similar response at the transcriptional level in the shared signaling pathways with genetic germline alterations in CVID. We aimed to explore the genomic substrate shared between these entities to better characterize the CVID phenotype immunodeficiency in NHL. By means of an in-silico approach, we interrogated the large, publicly available datasets contained in cBioPortal for the presence of genes associated with genetic pathogenic variants in a panel of 50 genes recurrently altered in CVID and previously described as causative or disease-modifying. We found that 323 (25%) of the 1,309 NHL samples available for analysis harbored variants of the CVID spectrum, with the most recurrent alteration presented in NHL occurring in PIK3CD (6%) and STAT3 (4%). Pathway analysis of common gene alterations showed enrichment in inflammatory, immune surveillance, and defective DNA repair mechanisms similar to those affected in CVID, with PIK3R1 appearing as a central node in the protein interaction network. The co-occurrence of gene alterations was a frequent phenomenon. This study represents an attempt to identify common genomic grounds between CVID and NHL. Further prospective studies are required to better know the role of genetic variants associated with CVID and their reflection on the somatic pathogenic variants responsible for cancer, as well as to characterize the CVID-like phenotype in NHL, with the potential to influence early CVID detection and therapeutic management.
Collapse
Affiliation(s)
- Kissy Guevara-Hoyer
- Cancer Immunomonitoring and Immuno-Mediated Pathologies Support Unit, IdSSC, Department of Clinical Immunology, San Carlos Clinical Hospital, Madrid, Spain
- Department of Clinical Immunology, IML and IdSSC, San Carlos Clinical Hospital, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | - Jesús Fuentes-Antrás
- Oncology Department, San Carlos Clinical Hospital, Madrid, Spain
- Experimental Therapeutics and Translational Oncology Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain
| | - Eduardo de la Fuente-Muñoz
- Cancer Immunomonitoring and Immuno-Mediated Pathologies Support Unit, IdSSC, Department of Clinical Immunology, San Carlos Clinical Hospital, Madrid, Spain
- Department of Clinical Immunology, IML and IdSSC, San Carlos Clinical Hospital, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | - Miguel Fernández-Arquero
- Cancer Immunomonitoring and Immuno-Mediated Pathologies Support Unit, IdSSC, Department of Clinical Immunology, San Carlos Clinical Hospital, Madrid, Spain
- Department of Clinical Immunology, IML and IdSSC, San Carlos Clinical Hospital, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | - Fernando Solano
- Department of Hematology, General University Hospital Nuestra Señora del Prado, Talavera de la Reina, Spain
| | | | - Esmeralda Neves
- Department of Immunology, Centro Hospitalar e Universitário do Porto, Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Hospital and University Center of Porto, Porto, Portugal
| | - Alberto Ocaña
- Oncology Department, San Carlos Clinical Hospital, Madrid, Spain
- Experimental Therapeutics and Translational Oncology Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain
| | - Rebeca Pérez de Diego
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, Madrid, Spain
| | - Silvia Sánchez-Ramón
- Cancer Immunomonitoring and Immuno-Mediated Pathologies Support Unit, IdSSC, Department of Clinical Immunology, San Carlos Clinical Hospital, Madrid, Spain
- Department of Clinical Immunology, IML and IdSSC, San Carlos Clinical Hospital, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
29
|
Savola P, Bhattacharya D, Huuhtanen J. The spectrum of somatic mutations in large granular lymphocyte leukemia, rheumatoid arthritis and Felty's syndrome. Semin Hematol 2022; 59:123-130. [DOI: 10.1053/j.seminhematol.2022.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/14/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022]
|
30
|
Zandi M, Moghaddam VA, Salehi Z, Mashayekhi F, Dalili S. The Impact of STAT3 rs1053005 Variation on Type 1 Diabetes Mellitus Susceptibility: Association Study and in Silico Analysis. Immunol Invest 2022; 51:1908-1919. [PMID: 35762640 DOI: 10.1080/08820139.2022.2079419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIMS Type 1 diabetes (T1DM) is an autoimmune disorder with multiple genetic and environmental risk factors that are still poorly understood. The signal transducer and activator of transcription (STAT) proteins play a pivotal role in immune-cell genesis and regulation. This study aimed to determine the effect of rs1053005 single nucleotide polymorphism (SNP) in 3'-UTR of STAT3 mRNA on the susceptibility to T1DM in an Iranian population. METHODS PCR-RFLP was conducted on 250 T1DM patients and 250 control cases to assess STAT3 rs1053005 polymorphism. Moreover, several bioinformatics tools were employed to identify the candidate miRNAs targeting the STAT3 mRNA region under study as well as the effect of rs1053005 on their binding site. RESULTS Significant variations in the distribution of genotypes and alleles were seen between cases and controls. The comparison results of the frequency of AA, AG, and GG genotypes between T1DM patients and control groups were 49.2% versus 64.8%, 39.2 versus 30%, and 11.6 versus 5.2%, respectively. Individuals who carried GG genotype were at 2.93-fold increased risk of developing T1DM and the G allele was associated with 1.79-fold higher T1DM risk. Bioinformatics analysis demonstrated that due to rs1053005, the interaction of 3 miRNAs were broken, 3 were weakened, 2 were reinforced, and 4 binding sites were created. CONCLUSION The result of this study indicates an association between STAT3 rs1053005 and T1DM susceptibility which may be due to interference of the SNP with native-binding site of some predicted miRNAs.
Collapse
Affiliation(s)
- Maryam Zandi
- Department of Biology, University Campus2, University of Guilan, Rasht, Iran
| | | | - Zivar Salehi
- Department of Biology, Faculty of sciences, University of Guilan, Rasht, Iran
| | - Farhad Mashayekhi
- Department of Biology, Faculty of sciences, University of Guilan, Rasht, Iran
| | - Setila Dalili
- Pediatric Diseases Research Center, Guilan University of medical sciences, Rasht, Iran
| |
Collapse
|
31
|
Lodi L, Faletti LE, Maccari ME, Consonni F, Groß M, Pagnini I, Ricci S, Heeg M, Simonini G, Azzari C, Ehl S. STAT3-confusion-of-function: beyond the loss and gain dualism. J Allergy Clin Immunol 2022; 150:1237-1241.e3. [PMID: 35750105 DOI: 10.1016/j.jaci.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Germline mutations of signal transducer and activator of transcription 3 (STAT3) are responsible for two distinct human diseases: autosomal-dominant hyper-immunoglobulin E syndrome (AD-HIES) caused by STAT3 loss-of-function (STAT3-LOF) mutations and STAT3 gain-of-function (STAT3-GOF) disease. So far, these entities have been regarded as antithetic, with AD-HIES mainly associated with characteristic infections and a connective tissue phenotype and STAT3-GOF characterized by lymphoproliferation and poly-autoimmunity. The R335W substitution in the DNA binding domain of STAT3 was initially described in 2 patients with typical AD-HIES, but paradoxically, recent functional analysis demonstrated a GOF effect of this variant. OBJECTIVE We describe a patient with Sjögren syndrome and features of AD-HIES with this mutation and further characterize its molecular consequences. METHODS We provide a clinical and immunological description of the patient. We studied STAT phosphorylation in primary patient cells and used A4 cells transfected with the patient allele to study phosphorylation kinetics, transcriptional activity and target-gene induction. RESULTS The hybrid clinical features of the patient were associated with normal Th17 cells. We observed enhanced and prolonged STAT3 phosphorylation, an increased STAT3 driven luciferase reporter activity upon interleukin-6 stimulation, but reduced IL-6 induced SOCS3 production. CONCLUSION The germline R335W-STAT3 variant displays a mixed behavior in vitro that mainly shows gain-of-function, but also loss-of-function features. This is matched by an ambiguous clinical and immunological phenotype which dismantles the classical antithetic dualism of gain- versus loss-of-function. Germline STAT3 mutation related-disease represents a pathological spectrum with the p.R335W associated phenotype locating between the two recognized clinical disease patterns.
Collapse
Affiliation(s)
- Lorenzo Lodi
- Department of Health Sciences, University of Florence, Florence, Italy; Immunology Unit, Department of Pediatrics, Meyer Children's Hospital, Florence, Italy
| | - Laura Eva Faletti
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Maria Elena Maccari
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Filippo Consonni
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Miriam Groß
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Ilaria Pagnini
- Rheumatology Unit, Department of Pediatrics, Meyer Children's University Hospital, Florence, Italy
| | - Silvia Ricci
- Department of Health Sciences, University of Florence, Florence, Italy; Immunology Unit, Department of Pediatrics, Meyer Children's Hospital, Florence, Italy
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Gabriele Simonini
- Rheumatology Unit, Department of Pediatrics, Meyer Children's University Hospital, Florence, Italy; NEUROFARBA Department, University of Florence, Florence, Italy
| | - Chiara Azzari
- Department of Health Sciences, University of Florence, Florence, Italy; Immunology Unit, Department of Pediatrics, Meyer Children's Hospital, Florence, Italy
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
32
|
Lobo PB, Guisado-Hernández P, Villaoslada I, de Felipe B, Carreras C, Rodriguez H, Carazo-Gallego B, Méndez-Echevarria A, Lucena JM, Aljaro PO, Castro MJ, Noguera-Uclés JF, Milner JD, McCann K, Zimmerman O, Freeman AF, Lionakis MS, Holland SM, Neth O, Olbrich P. Ex vivo effect of JAK inhibition on JAK-STAT1 pathway hyperactivation in patients with dominant-negative STAT3 mutations. J Clin Immunol 2022; 42:1193-1204. [PMID: 35507130 DOI: 10.1007/s10875-022-01273-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE STAT1 gain-of-function (GOF) and dominant-negative (DN) STAT3 syndromes share clinical manifestations including infectious and inflammatory manifestations. Targeted treatment with Janus-kinase (JAK) inhibitors shows promising results in treating STAT1 GOF-associated symptoms while management of DN STAT3 patients has been largely supportive. We here assessed the impact of ruxolitinib on the JAK-STAT1/3 pathway in DN STAT3 patients' cells. METHODS Using flow cytometry, immunoblot, qPCR, and ELISA techniques, we examined the levels of basal STAT1 and phosphorylated STAT1 (pSTAT1) of cells obtained from DN STAT3, STAT1 GOF patients, and healthy donors following stimulation with type I/II interferons (IFNs) or interleukin (IL)-6. We also describe the impact of ruxolitinib on cytokine-induced STAT1 signaling in these patients. RESULTS DN STAT3 and STAT1 GOF resulted in a similar phenotype characterized by increased STAT1 and pSTAT1 levels in response to IFNα (CD3+ cells) and IFNγ (CD14+ monocytes). STAT1-downstream gene expression and C-X-C motif chemokine 10 secretion were higher in most DN STAT3 patients upon stimulation compared to healthy controls. Ex vivo treatment with the JAK1/2-inhibitor ruxolitinib reduced cytokine responsiveness and normalized STAT1 phosphorylation in DN STAT3 and STAT1 GOF patient' cells. In addition, ex vivo treatment was effective in modulating STAT1 downstream signaling in DN STAT3 patients. CONCLUSION In the absence of effective targeted treatment options for AD-HIES at present, modulation of the JAK/STAT1 pathway with JAK inhibitors may be further explored particularly in those AD-HIES patients with autoimmune and/or autoinflammatory manifestations.
Collapse
Affiliation(s)
- Pilar Blanco Lobo
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocío, Institute of Biomedicine of Seville (IBIS)/Universidad de Sevilla/CSIC, Red de Investigación Traslacional en Infectología Pediátrica RITIP, Av Manuel Siurot s/n, 41013, Seville, Spain
| | - Paloma Guisado-Hernández
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocío, Institute of Biomedicine of Seville (IBIS)/Universidad de Sevilla/CSIC, Red de Investigación Traslacional en Infectología Pediátrica RITIP, Av Manuel Siurot s/n, 41013, Seville, Spain
| | - Isabel Villaoslada
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocío, Institute of Biomedicine of Seville (IBIS)/Universidad de Sevilla/CSIC, Red de Investigación Traslacional en Infectología Pediátrica RITIP, Av Manuel Siurot s/n, 41013, Seville, Spain
| | - Beatriz de Felipe
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocío, Institute of Biomedicine of Seville (IBIS)/Universidad de Sevilla/CSIC, Red de Investigación Traslacional en Infectología Pediátrica RITIP, Av Manuel Siurot s/n, 41013, Seville, Spain
| | - Carmen Carreras
- Pediatric Infectious Diseases and Immunodeficiency Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Hector Rodriguez
- Pediatric Infectious Diseases and Immunodeficiency Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Begoña Carazo-Gallego
- Pediatric Infectology and Immunodeficiencies Unit, IBIMA, Department of Pediatrics, Hospital Regional Universitario Málaga, Malaga, Spain
| | - Ana Méndez-Echevarria
- Pediatric Infectious and Tropical Diseases Department, Hospital Universitario La Paz, CIBERINFEC, Carlos III Health Institute, Madrid, Spain
| | | | | | - María José Castro
- Servicio de Citometría y Separación Celular, Instituto de Biomedicina de Sevilla - IBiS/HUVR/US/CSIC, Seville, Spain
| | | | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Katelyn McCann
- Laboratory of Clinical Immunology and Microbiology, Immunopathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ofer Zimmerman
- Department of Medicine, Division of Allergy/Immunology, Washington University in St Louis, St Louis, MO, USA
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, Immunopathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, LCIM, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, Immunopathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olaf Neth
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocío, Institute of Biomedicine of Seville (IBIS)/Universidad de Sevilla/CSIC, Red de Investigación Traslacional en Infectología Pediátrica RITIP, Av Manuel Siurot s/n, 41013, Seville, Spain.
| | - Peter Olbrich
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocío, Institute of Biomedicine of Seville (IBIS)/Universidad de Sevilla/CSIC, Red de Investigación Traslacional en Infectología Pediátrica RITIP, Av Manuel Siurot s/n, 41013, Seville, Spain
| |
Collapse
|
33
|
Wu J, Jin YY, Gong RL, Yang F, Su XY, Chen TX. Genome-Wide ChIP-seq and RNA-seq Analyses of STAT3 Target Genes in TLRs Activated Human Peripheral Blood B Cells. Front Immunol 2022; 13:821457. [PMID: 35345674 PMCID: PMC8957201 DOI: 10.3389/fimmu.2022.821457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/10/2022] [Indexed: 11/29/2022] Open
Abstract
Toll like receptors (TLRs) induced response plays a vital role in B-cell development and activation, in which TLR7-mediated and TLR9-mediated response interact together and play antagonistic or cooperative roles at different situations. Previous studies showed that the transcription factor signal transducer and activator of transcription (STAT) 3 was one of the key transcriptional factors (TFs) needed for both TLR7 and TLR9 signaling in B cell, and patients with autosomal dominant hyper IgE syndromes (AD-HIES) due to STAT3 mutations having defective TLRs response in B cells. However, how STAT3 affects its target genes and the downstream signaling pathways in B cell upon TLRs stimulation remains unclarified on a genome-wide level. ChIP-seq and RNA-seq was used in this study to identify the STAT3 targets in response to TLRs stimulation in human B cell. STAT3 ChIP-seq results showed a total of 611 and 2,289 differential STAT3-binding sites in human B cell after TLR7 and TLR9 agonists stimulation, respectively. RNA-seq results showed 1,186 and 1,775 differentially expressed genes after TLR7 and TLR9 activation, respectively. We identified 47 primary STAT3 target genes after TLR7 activation and 189 target genes after TLR9 activation in B cell by integration of STAT3 ChIP-seq and RNA-seq data. Among these STAT3 primary targets, we identified 7 TFs and 18 TFs for TLR7 and TLR9 response, respectively. Besides, we showed that STAT3 might regulate TLR9, but not TLR7 response in B cells through directly regulating integrin signaling pathway, which might further affect the antagonism between TLR7 and TLR9 signaling in B cell. Our study provides insights into the molecular mechanism of human TLRs response in B cell and how it can be regulated, which helps to better understand and modulate TLR-mediated pathogenic immune responses in B cell.
Collapse
Affiliation(s)
- Jing Wu
- Division of Immunology, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Allergy/Immunology Innovation Team, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Ying Jin
- Allergy/Immunology Innovation Team, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology/Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruo-Lan Gong
- Division of Immunology, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Yang
- Division of Immunology, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Ya Su
- Division of Immunology, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tong-Xin Chen
- Division of Immunology, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Allergy/Immunology Innovation Team, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology/Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
34
|
Cekic S, Huriyet H, Hortoglu M, Kasap N, Ozen A, Karakoc-Aydiner E, Metin A, Ocakoglu G, Demiroz Abakay C, Temel SG, Ozemri Sag S, Baris S, Cavas T, Kilic SS. Full Increased radiosensitivity and impaired DNA repair in patients with STAT3-LOF and ZNF341 deficiency, potentially contributing to malignant transformations. Clin Exp Immunol 2022; 209:83-89. [PMID: 35511492 PMCID: PMC9307231 DOI: 10.1093/cei/uxac041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 04/03/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
STAT3 plays an important role in various complex and sometimes contradictory pathways such as proliferation, differentiation, migration, inflammation, and apoptosis. The transcriptional activity of the STAT3 gene is controlled by a transcription factor called ZNF341. There is insufficient data on radiation sensitivity and post-radiation DNA repair in STAT3- loss-of-function (LOF) patients. We aimed to investigate the radiosensitivity in patients with STAT3-LOF and ZNF341 deficiency. Twelve patients with STAT3-LOF and four ZNF341-deficiency patients were recruited from three clinical immunology centers in Turkey and evaluated for radiosensitivity by the Comet assay, comparing to 14 age- and sex-matched healthy controls. The Tail length (μm), Tail DNA (%), Olive Tail Moment (OTM) (arbitrary units) were evaluated at the same time for baseline (spontaneous), initial (immediately after 2Gy irradiation), and recovery (2h after irradiation) periods by using a computerized image-analysis system, estimating DNA damage. Except for a patient with ZNF341 deficiency who developed nasal cell primitive neuroendocrine tumor and papillary thyroid cancer during the follow-up, there was no cancer in both groups. During the recovery period of irradiation, TL, TDNA%, and OTM values of healthy controls decreased rapidly towards the baseline, while these values of patients with STAT3-LOF and ZNF341 deficiency continued to increase, implying impaired DNA repair mechanisms. Increased radiosensitivity and impaired DNA repair were demonstrated in patients diagnosed with STAT3-LOF and ZNF341 deficiency, potentially explaining the susceptibility to malignant transformation.
Collapse
Affiliation(s)
- Sukru Cekic
- Division of Pediatric Immunology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - Huzeyfe Huriyet
- Faculty of Sciences and Letters, Biology Department, Uludag University, Bursa, Turkey
| | - Melika Hortoglu
- Faculty of Sciences and Letters, Biology Department, Uludag University, Bursa, Turkey
| | - Nurhan Kasap
- Division of Pediatric Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey
| | - Ahmet Ozen
- Division of Pediatric Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey.,The Isil Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Division of Pediatric Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey.,The Isil Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ayse Metin
- Division of Pediatric Allergy and Immunology, University of Health Sciences/Ankara City Hospital/Children's Hospital, Ankara, Turkey
| | - Gokhan Ocakoglu
- Department of Biostatistics, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - Candan Demiroz Abakay
- Department of Radiation Oncology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Sehime G Temel
- Department of Medical Genetics, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - Sebnem Ozemri Sag
- Department of Medical Genetics, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - Safa Baris
- Division of Pediatric Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey.,The Isil Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Tolga Cavas
- Faculty of Sciences and Letters, Biology Department, Uludag University, Bursa, Turkey
| | - Sara Sebnem Kilic
- Division of Pediatric Immunology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey.,Bursa Uludag University, Translational Medicine, Bursa, Turkey
| |
Collapse
|
35
|
Cortesi M, Soresina A, Dotta L, Gorio C, Cattalini M, Lougaris V, Porta F, Badolato R. Pathogenesis of Autoimmune Cytopenias in Inborn Errors of Immunity Revealing Novel Therapeutic Targets. Front Immunol 2022; 13:846660. [PMID: 35464467 PMCID: PMC9019165 DOI: 10.3389/fimmu.2022.846660] [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: 12/31/2021] [Accepted: 03/21/2022] [Indexed: 12/02/2022] Open
Abstract
Autoimmune diseases are usually associated with environmental triggers and genetic predisposition. However, a few number of autoimmune diseases has a monogenic cause, mostly in children. These diseases may be the expression, isolated or associated with other symptoms, of an underlying inborn error of immunity (IEI). Autoimmune cytopenias (AICs), including immune thrombocytopenic purpura (ITP), autoimmune hemolytic anemia (AIHA), autoimmune neutropenia (AN), and Evans’ syndrome (ES) are common presentations of immunological diseases in the pediatric age, with at least 65% of cases of ES genetically determined. Autoimmune cytopenias in IEI have often a more severe, chronic, and relapsing course. Treatment refractoriness also characterizes autoimmune cytopenia with a monogenic cause, such as IEI. The mechanisms underlying autoimmune cytopenias in IEI include cellular or humoral autoimmunity, immune dysregulation in cases of hemophagocytosis or lymphoproliferation with or without splenic sequestration, bone marrow failure, myelodysplasia, or secondary myelosuppression. Genetic characterization of autoimmune cytopenias is of fundamental importance as an early diagnosis improves the outcome and allows the setting up of a targeted therapy, such as CTLA-4 IgG fusion protein (Abatacept), small molecule inhibitors (JAK-inhibitors), or gene therapy. Currently, gene therapy represents one of the most attractive targeted therapeutic approaches to treat selected inborn errors of immunity. Even in the absence of specific targeted therapies, however, whole exome genetic testing (WES) for children with chronic multilineage cytopenias should be considered as an early diagnostic tool for disease diagnosis and genetic counseling.
Collapse
Affiliation(s)
- Manuela Cortesi
- Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, ASST- Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Annarosa Soresina
- Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, ASST- Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Laura Dotta
- Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, ASST- Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Chiara Gorio
- Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, ASST- Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Marco Cattalini
- Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, ASST- Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Vassilios Lougaris
- Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, ASST- Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Fulvio Porta
- Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, ASST- Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Raffaele Badolato
- Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, ASST- Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| |
Collapse
|
36
|
Staller MV, Ramirez E, Kotha SR, Holehouse AS, Pappu RV, Cohen BA. Directed mutational scanning reveals a balance between acidic and hydrophobic residues in strong human activation domains. Cell Syst 2022; 13:334-345.e5. [PMID: 35120642 PMCID: PMC9241528 DOI: 10.1016/j.cels.2022.01.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/20/2021] [Accepted: 01/05/2022] [Indexed: 01/01/2023]
Abstract
Acidic activation domains are intrinsically disordered regions of the transcription factors that bind coactivators. The intrinsic disorder and low evolutionary conservation of activation domains have made it difficult to identify the sequence features that control activity. To address this problem, we designed thousands of variants in seven acidic activation domains and measured their activities with a high-throughput assay in human cell culture. We found that strong activation domain activity requires a balance between the number of acidic residues and aromatic and leucine residues. These findings motivated a predictor of acidic activation domains that scans the human proteome for clusters of aromatic and leucine residues embedded in regions of high acidity. This predictor identifies known activation domains and accurately predicts previously unidentified ones. Our results support a flexible acidic exposure model of activation domains in which the acidic residues solubilize hydrophobic motifs so that they can interact with coactivators. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Max V Staller
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA; Center for Computational Biology, University of California Berkeley, Berkeley, CA 94720, USA.
| | - Eddie Ramirez
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Sanjana R Kotha
- Center for Computational Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA; Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Rohit V Pappu
- Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Barak A Cohen
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA.
| |
Collapse
|
37
|
Cording S, Lhermitte L, Malamut G, Berrabah S, Trinquand A, Guegan N, Villarese P, Kaltenbach S, Meresse B, Khater S, Dussiot M, Bras M, Cheminant M, Tesson B, Bole-Feysot C, Bruneau J, Molina TJ, Sibon D, Macintyre E, Hermine O, Cellier C, Asnafi V, Cerf-Bensussan N. Oncogenetic landscape of lymphomagenesis in coeliac disease. Gut 2022; 71:497-508. [PMID: 33579790 PMCID: PMC8862029 DOI: 10.1136/gutjnl-2020-322935] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Enteropathy-associated T-cell lymphoma (EATL) is a rare but severe complication of coeliac disease (CeD), often preceded by low-grade clonal intraepithelial lymphoproliferation, referred to as type II refractory CeD (RCDII). Knowledge on underlying oncogenic mechanisms remains scarce. Here, we analysed and compared the mutational landscape of RCDII and EATL in order to identify genetic drivers of CeD-associated lymphomagenesis. DESIGN Pure populations of RCDII-cells derived from intestinal biopsies (n=9) or sorted from blood (n=2) were analysed by whole exome sequencing, comparative genomic hybridisation and RNA sequencing. Biopsies from RCDII (n=50), EATL (n=19), type I refractory CeD (n=7) and uncomplicated CeD (n=18) were analysed by targeted next-generation sequencing. Moreover, functional in vitro studies and drug testing were performed in RCDII-derived cell lines. RESULTS 80% of RCDII and 90% of EATL displayed somatic gain-of-functions mutations in the JAK1-STAT3 pathway, including a remarkable p.G1097 hotspot mutation in the JAK1 kinase domain in approximately 50% of cases. Other recurrent somatic events were deleterious mutations in nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) regulators TNFAIP3 and TNIP3 and potentially oncogenic mutations in TET2, KMT2D and DDX3X. JAK1 inhibitors, and the proteasome inhibitor bortezomib could block survival and proliferation of malignant RCDII-cell lines. CONCLUSION Mutations activating the JAK1-STAT3 pathway appear to be the main drivers of CeD-associated lymphomagenesis. In concert with mutations in negative regulators of NF-κB, they may favour the clonal emergence of malignant lymphocytes in the cytokine-rich coeliac intestine. The identified mutations are attractive therapeutic targets to treat RCDII and block progression towards EATL.
Collapse
Affiliation(s)
- Sascha Cording
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France
| | - Ludovic Lhermitte
- Université de Paris, Institut Necker-Enfants Malades, INSERM UMR 1151, Paris, France,Laboratory of Onco-Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - Georgia Malamut
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France,Department of Gastroenterology, AP-HP, Hôpital Cochin, Paris, France
| | - Sofia Berrabah
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France
| | - Amélie Trinquand
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France,Haematology Department, National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Nicolas Guegan
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France
| | - Patrick Villarese
- Université de Paris, Institut Necker-Enfants Malades, INSERM UMR 1151, Paris, France,Laboratory of Onco-Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - Sophie Kaltenbach
- Department of Cytogenetics, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - Bertrand Meresse
- Université de Lille, CHU Lille, INSERM UMR 1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
| | - Sherine Khater
- Department of Gastroenterology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Michael Dussiot
- Université de Paris, Imagine Institute, Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM UMR 1163, Paris, France
| | - Marc Bras
- Université de Paris, Imagine Institute, Bioinformatics Platform, Paris, France
| | - Morgane Cheminant
- Université de Paris, Imagine Institute, Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM UMR 1163, Paris, France,Clinical Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | | | | | - Julie Bruneau
- Department of Pathology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - Thierry Jo Molina
- Université de Paris, Imagine Institute, Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM UMR 1163, Paris, France,Department of Pathology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - David Sibon
- Clinical Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - Elizabeth Macintyre
- Université de Paris, Institut Necker-Enfants Malades, INSERM UMR 1151, Paris, France,Laboratory of Onco-Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - Olivier Hermine
- Université de Paris, Imagine Institute, Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM UMR 1163, Paris, France,Clinical Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - Christophe Cellier
- Department of Gastroenterology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Vahid Asnafi
- Université de Paris, Institut Necker-Enfants Malades, INSERM UMR 1151, Paris, France,Laboratory of Onco-Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France
| | - Nadine Cerf-Bensussan
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France
| | | |
Collapse
|
38
|
Araki T, Watanabe Y, Okada Y, Murakami H, Ogo N, Asai A. Identification of serum and glucocorticoid-regulated kinase 1 as a regulator of signal transducer and activator of transcription 3 signaling. Exp Cell Res 2022; 413:113079. [PMID: 35202674 DOI: 10.1016/j.yexcr.2022.113079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/20/2021] [Accepted: 02/11/2022] [Indexed: 11/18/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays key roles in cancer cell proliferation, invasion, and immunosuppression. In many human cancer cells, STAT3 is hyperactivated, which leads to tumor progression and drug resistance, and therefore STAT3 and its modulators are considered effective drug targets. However, the complex regulatory mechanisms of STAT3 have made it difficult to develop potent anticancer drugs that suppress its activity. Here, we report serum and glucocorticoid-regulated kinase 1 (SGK1) as a novel regulator of STAT3 signaling and an effective target for combination therapy with Janus kinase (JAK) inhibitors. We screened small molecules using a gain-of-function mutant of STAT3 resistant to JAK inhibition and found that an SGK1 inhibitor suppressed the constitutive activation of STAT3. Importantly, our results revealed that SGK1 also mediated the activation of wild-type STAT3. Further examination suggested that the tuberous sclerosis complex 2 and mammalian target of rapamycin signaling pathway were involved in STAT3 activation by SGK1. Finally, we demonstrated that SGK1 inhibition enhanced the inhibitory effect of a JAK inhibitor on STAT3 phosphorylation and cancer cell proliferation. Our findings provide new insights into the molecular mechanisms of STAT3 activation and suggest SGK1 as a potential target for STAT3-targeted combination cancer therapy.
Collapse
Affiliation(s)
- Toshihiro Araki
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan; Discovery Technology Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Muraoka-Higashi, Fujisawa, Kanagawa, Japan
| | - Yuuki Watanabe
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan; Public Affairs and Policy Department, Mitsubishi Tanabe Pharma Corporation, Marunouchi, Chiyoda-ku, Tokyo, Japan
| | - Yusuke Okada
- Sohyaku Project Planning & Management Department, Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Marunouchi, Chiyoda-ku, Tokyo, Japan
| | - Hisashi Murakami
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan.
| |
Collapse
|
39
|
Jouhault Q, Cherqaoui B, Jobart-Malfait A, Glatigny S, Lauraine M, Hulot A, Morelle G, Hagege B, Ermoza K, El Marjou A, Izac B, Saintpierre B, Letourneur F, Rémy S, Anegon I, Boissier MC, Chiocchia G, Breban M, Araujo LM. Interleukin 27 is a novel cytokine with anti-inflammatory effects against spondyloarthritis through the suppression of Th17 responses. Front Immunol 2022; 13:1072420. [PMID: 36818477 PMCID: PMC9933703 DOI: 10.3389/fimmu.2022.1072420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/19/2022] [Indexed: 02/05/2023] Open
Abstract
Introduction Spondylarthritis (SpA) development in HLA-B27/human β2-microglobulin transgenic rat (B27-rat) is correlated with altered conventional dendritic cell (cDC) function that promotes an inflammatory pattern of CD4+T cells, including a biased expansion of pro-inflammatory Th17 population and imbalance of regulatory T cells cytokine profile. Transcriptomic analysis revealed that cDCs from B27-rats under express IL-27, an anti-inflammatory cytokine which induces the differentiation of IL-10+ regulatory T cells and inhibits Th17 cells. Methods Here, we first investigated whether in vitro addition of exogenous IL-27 could reverse the inflammatory pattern observed in CD4+ T cells. Next, we performed preclinical assay using IL-27 to investigate whether in vivo treatment could prevent SpA development in B27-rats. Results in vitro addition of IL-27 to cocultures of cDCs and CD4+ T cell subsets from B27-rats reduced IL-17 and enhanced IL-10 production by T cells. Likewise, IL-27 inhibited the production of IL-17 by CD4+ T cells from SpA patients. Interestingly, in vivo treatment with recombinant IL-27 starting before SpA onset, inhibited SpA development in B27-rats through the suppression of IL-17/TNF producing CD4+ T cells. Discussion Overall, our results reveal a potent inhibitory effect of IL-27 and highlight this cytokine as a promising new therapeutic target in SpA, especially for SpA patients non responders to currently approved biotherapies.
Collapse
Affiliation(s)
- Quentin Jouhault
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Bilade Cherqaoui
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Aude Jobart-Malfait
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Simon Glatigny
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Marc Lauraine
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Audrey Hulot
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Guillaume Morelle
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Benjamin Hagege
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Kétia Ermoza
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Ahmed El Marjou
- Plateforme de production d'anticorps et de protéines recombinantes-Institut Curie/CNRS UMR144, Paris, France
| | - Brigitte Izac
- Plateforme GenomIC- Université de Paris, Institut Cochin, INSERM-CNRS, Paris, France
| | - Benjamin Saintpierre
- Plateforme GenomIC- Université de Paris, Institut Cochin, INSERM-CNRS, Paris, France
| | - Franck Letourneur
- Plateforme GenomIC- Université de Paris, Institut Cochin, INSERM-CNRS, Paris, France
| | - Séverine Rémy
- Platform Transgenic Rats and ImmunoPhenomics, INSERM UMR 1064-CRTI, Nantes, France
| | - Ignacio Anegon
- Platform Transgenic Rats and ImmunoPhenomics, INSERM UMR 1064-CRTI, Nantes, France
| | - Marie-Christophe Boissier
- Inserm UMR1125-Université Sorbonne Paris Nord, Rheumatology Division, Avicenne Hospital (AP-HP), Bobigny, France
| | - Gilles Chiocchia
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France.,Haematology-Immunology Division, Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France
| | - Maxime Breban
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France.,Rheumatology Division, Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France
| | - Luiza M Araujo
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris-Centre, Paris, France
| |
Collapse
|
40
|
STAT3 is critical for skeletal development and bone homeostasis by regulating osteogenesis. Nat Commun 2021; 12:6891. [PMID: 34824272 PMCID: PMC8616950 DOI: 10.1038/s41467-021-27273-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 10/19/2021] [Indexed: 11/08/2022] Open
Abstract
Skeletal deformities are typical AD-HIES manifestations, which are mainly caused by heterozygous and loss-of-function mutations in Signal transducer and activator of transcription 3 (STAT3). However, the mechanism is still unclear and the treatment strategy is limited. Herein, we reported that the mice with Stat3 deletion in osteoblasts, but not in osteoclasts, induced AD-HIES-like skeletal defects, including craniofacial malformation, osteoporosis, and spontaneous bone fracture. Mechanistic analyses revealed that STAT3 in cooperation with Msh homeobox 1(MSX1) drove osteoblast differentiation by promoting Distal-less homeobox 5(Dlx5) transcription. Furthermore, pharmacological activation of STAT3 partially rescued skeletal deformities in heterozygous knockout mice, while inhibition of STAT3 aggravated bone loss. Taken together, these data show that STAT3 is critical for modulating skeletal development and maintaining bone homeostasis through STAT3-indcued osteogenesis and suggest it may be a potential target for treatments.
Collapse
|
41
|
Costagliola G, Cappelli S, Consolini R. Autoimmunity in Primary Immunodeficiency Disorders: An Updated Review on Pathogenic and Clinical Implications. J Clin Med 2021; 10:jcm10204729. [PMID: 34682853 PMCID: PMC8538991 DOI: 10.3390/jcm10204729] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022] Open
Abstract
During the last years, studies investigating the intriguing association between immunodeficiency and autoimmunity led to the discovery of new monogenic disorders, the improvement in the knowledge of the pathogenesis of autoimmunity, and the introduction of targeted treatments. Autoimmunity is observed with particular frequency in patients with primary antibody deficiencies, such as common variable immunodeficiency (CVID) and selective IgA deficiency, but combined immunodeficiency disorders (CIDs) and disorders of innate immunity have also been associated with autoimmunity. Among CIDs, the highest incidence of autoimmunity is described in patients with autoimmune polyendocrine syndrome 1, LRBA, and CTLA-4 deficiency, and in patients with STAT-related disorders. The pathogenesis of autoimmunity in patients with immunodeficiency is far to be fully elucidated. However, altered germ center reactions, impaired central and peripheral lymphocyte negative selection, uncontrolled lymphocyte proliferation, ineffective cytoskeletal function, innate immune defects, and defective clearance of the infectious agents play an important role. In this paper, we review the main immunodeficiencies associated with autoimmunity, focusing on the pathogenic mechanisms responsible for autoimmunity in each condition and on the therapeutic strategies. Moreover, we provide a diagnostic algorithm for the diagnosis of PIDs in patients with autoimmunity.
Collapse
|
42
|
Guo ZS, Qu Z. PDLIM2: Signaling pathways and functions in cancer suppression and host immunity. Biochim Biophys Acta Rev Cancer 2021; 1876:188630. [PMID: 34571051 DOI: 10.1016/j.bbcan.2021.188630] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/30/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022]
Abstract
PDZ and LIM domains-containing proteins play pivotal functions in cell cytoskeleton organization, cell polarization and differentiation. As a key member of the family, PDLIM2 regulates stability and activity of transcription factors such as NF-κB, STATs and β-catenin, and thus exert it functions in inflammation, immunity, and cancer. PDLIM2 functions as a tumor suppressor in multiple tissues and it is often genetically mutated or epigenetically silenced in human cancers derived from lung, breast, ovarian and other histologies. However, in certain types of cancers, PDLIM2 may promote cancer cell proliferation and metastases. Therefore, PDLIM2 is added to a long list of genes that can function as tumor suppressor or oncogenic protein. During tumorigenesis induced by oncogenic viruses, PDLIM2 is a key target. Through promotion of NF-κB/RelA and STAT3 degradation, PDLIM2 enhances expression of proteins involved in antigen presentation and promotes T-cell activation while repressing multidrug resistance genes, thereby rendering mutated cells susceptible to immune surveillance and cytotoxicity mediated by immune cells and chemotherapeutic drugs. Intriguingly, PDLIM2 in alveolar macrophages (AMs) plays key roles in monitoring lung tumorigenesis, as its selective genetic deletion leads to constitutive activation of STAT3, driving monocyte differentiation to AMs with pro-tumorigenic polarization and activation. PDLIM2 has also been explored as a therapeutic target for cancer therapy. At the end of this review, we provide perspectives on this important molecule and discuss the future directions of both basic and translational studies.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Zhaoxia Qu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
43
|
Kattner AA. We refuse to die - T cells causing havoc. Biomed J 2021; 44:377-382. [PMID: 34508914 PMCID: PMC8514847 DOI: 10.1016/j.bj.2021.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 10/28/2022] Open
Abstract
This issue of the Biomedical Journal offers insights into the origin and consequences of different lymphoproliferative disorders and autoimmunity. Furthermore we learn about RASopathies, a group of congenital disorders that occur rather frequently. Then the current ELISA assays for measuring antibody avidity are critically examined, the relationship between female sex steroid hormones and cardiovascular disease is explored, and an assessment of persistent diarrhea as a leading cause of child death in India is performed. Additionally, there are several articles about COVID-19, presenting its connection to neutrophil recruitment and acute respiratory distress syndrome, as well as its relation to changes in the vascular glycocalyx. A COVID-19 case study from the emergency room is presented. We are also introduced to novel treatment approaches against COVID-19 like the construction of peptide-based vaccines, or targeting the respiratory tract microbiome. Finally, there is an assessment about how prepared medical students at a Taiwan University feel for independent practice, and another article about the treatment of intravascular large B cell lymphoma in a Taiwanese institution. Lastly, we discover possible surgery techniques in the case of external auditory canal osteoma.
Collapse
|
44
|
Intestinal immunoregulation: lessons from human mendelian diseases. Mucosal Immunol 2021; 14:1017-1037. [PMID: 33859369 DOI: 10.1038/s41385-021-00398-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023]
Abstract
The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.
Collapse
|
45
|
Zeng L, Li A, Zhang Z, Zhang F, Chen H, Wang Y, Ding X, Luo H. Ropivacaine Induces Cell Cycle Arrest in the G0/G1 Phase and Apoptosis of PC12 Cells via Inhibiting Mitochondrial STAT3 Translocation. Inflammation 2021; 44:2362-2376. [PMID: 34417665 DOI: 10.1007/s10753-021-01508-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/24/2021] [Indexed: 11/25/2022]
Abstract
STAT3 has neuroprotective effect via non-canonical activation and mitochondrial translocation, but its effect on ropivacaine-induced neurotoxicity remains unclear. Our previous study revealed that apoptosis was an important mechanism of ropivacaine-induced neurotoxicity; this study is to illustrate the relationship between STAT3 with ropivacaine-induced apoptosis. Those results showed that ropivacaine treatment decreased cell viability, induced cell cycle arrest in the G0/G1 phase, apoptosis, oxidative stress, and mitochondrial dysfunction in PC12 cells. Moreover, ropivacaine decreased the phosphorylated levels of STAT3 at Ser727 and downregulated the expression of STAT3 upstream gene IL-6. The mitochondrial translocation of STAT3 was also hindered by ropivacaine. To further illustrate the connection of STAT3 protein structure with ropivacaine, the autodock-vina was used to examine the interaction between STAT3 and ropivacaine, and the results showed that ropivacaine could bind to STAT3's proline site and other sites. In addition, the activator and inhibitor of mitoSTAT3 translocation were used to demonstrate it was involved in ropivacaine-induced apoptosis; the results showed that enhancing the mitochondrial STAT3 translocation could prevent ropivacaine-induced apoptosis. Finally, the expression of p-STAT3 and the levels of apoptosis in the spinal cord were also detected; the results were consistent with the cell experiment; ropivacaine decreased the expression of p-STAT3 protein and increased the levels of apoptosis in the spinal cord. We demonstrated that ropivacaine induced apoptosis by inhibiting the phosphorylation of STAT3 at Ser727 and the mitochondrial STAT3 translocation. This effect was reversed by the activation of the mitochondrial STAT3 translocation.
Collapse
Affiliation(s)
- Lian Zeng
- Department of Anesthesiology, Xiangyang Key Laboratory of Movement Disorders, Xiangyang No.1 People's Hospital, Hubei Clinical Research Center of Parkinson's Disease, Hubei University of Medicine, Hubei, China
| | - Aohan Li
- Department of Anesthesiology, Xiangyang Key Laboratory of Movement Disorders, Xiangyang No.1 People's Hospital, Hubei Clinical Research Center of Parkinson's Disease, Hubei University of Medicine, Hubei, China
| | - Zhen Zhang
- Department of Anesthesiology, Xiangyang Key Laboratory of Movement Disorders, Xiangyang No.1 People's Hospital, Hubei Clinical Research Center of Parkinson's Disease, Hubei University of Medicine, Hubei, China
| | - Fuyu Zhang
- Department of Anesthesiology, Xiangyang Key Laboratory of Movement Disorders, Xiangyang No.1 People's Hospital, Hubei Clinical Research Center of Parkinson's Disease, Hubei University of Medicine, Hubei, China
| | - Huaxian Chen
- Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Hubei, China
| | - Ying Wang
- Department of Anesthesiology, Xiangyang Key Laboratory of Movement Disorders, Xiangyang No.1 People's Hospital, Hubei Clinical Research Center of Parkinson's Disease, Hubei University of Medicine, Hubei, China
| | - Xudong Ding
- Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Hubei, China
| | - Huiyu Luo
- Department of Anesthesiology, Xiangyang Key Laboratory of Movement Disorders, Xiangyang No.1 People's Hospital, Hubei Clinical Research Center of Parkinson's Disease, Hubei University of Medicine, Hubei, China.
- Department of Rehabilitation Medicine, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Hubei, China.
| |
Collapse
|
46
|
Gargalionis AN, Papavassiliou KA, Papavassiliou AG. Targeting STAT3 Signaling Pathway in Colorectal Cancer. Biomedicines 2021; 9:1016. [PMID: 34440220 PMCID: PMC8392110 DOI: 10.3390/biomedicines9081016] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/31/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a critical transcription factor that has been firmly associated with colorectal cancer (CRC) initiation and development. STAT3 mediates key inflammatory mechanisms in colitis-associated cancer, becomes excessively activated in CRC, and enhances cancer cell proliferation, tumor growth, angiogenesis, invasion, and migration. STAT3 hyperactivation in malignant cells, surrounding immune cells and cancer-associated fibroblasts, mediates inhibition of the innate and adaptive immunity of the tumor microenvironment, and, therefore, tumor evasion from the immune system. These features highlight STAT3 as a promising therapeutic target; however, the mechanisms underlying these features have not been fully elucidated yet and STAT3 inhibitors have not reached the clinic in everyday practice. In the present article, we review the STAT3 signaling network in CRC and highlight the current notion for the design of STAT3-focused treatment approaches. We also discuss recent breakthroughs in combination immunotherapy regimens containing STAT3 inhibitors, therefore providing a new perception for the clinical application of STAT3 in CRC.
Collapse
Affiliation(s)
- Antonios N. Gargalionis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.N.G.); (K.A.P.)
- Department of Biopathology, Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Kostas A. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.N.G.); (K.A.P.)
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.N.G.); (K.A.P.)
| |
Collapse
|
47
|
Marega LF, Sabino JS, Pedroni MV, Teocchi M, Lanaro C, de Albuquerque DM, Dos Santos IP, Costa FF, Dos Santos Vilela MM. Phenotypes of STAT3 gain-of-function variant related to disruptive regulation of CXCL8/STAT3, KIT/STAT3, and IL-2/CD25/Treg axes. Immunol Res 2021; 69:445-456. [PMID: 34390446 DOI: 10.1007/s12026-021-09225-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/27/2021] [Indexed: 12/01/2022]
Abstract
STAT3 is a cytokine-signaling transcription factor critical for gene regulation. Gain-of-function (GOF) mutations in STAT3 are associated with lymphoproliferation, autoimmune cytopenias, increased susceptibility to infection, early-onset solid-organ autoimmunity, short stature, and eczema. We studied the JAK/STAT signaling pathway gene expression and the cytokine profile in two families carrying STAT3-GOF variants to shed light on the STAT3-GOF-associated variable expressivity, including the identification of disease markers. Considering 92 target genes, KIT and IL2RA were downregulated only in patients with high clinical penetrance, while CXCL8 was markedly downregulated for all of them. Unlike previous studies, SOCS3-a STAT3 inhibitor-was not upregulated in patients. In addition, low levels of IL-2 and a reduced numbers of Tregs cells were strikingly prevalent in patients. This study shows a disruptive role of STAT3-GOF variants in the regulatory axis activities CXCL8/STAT3, KIT/STAT3, IL2/CD25/Treg, which, by slightly different mechanisms, underlie the broad clinical spectrum seen in the studied patients. In addition, we suggest the investigation of CXCL8 as a biomarker for identifying STAT3-GOF mutation.
Collapse
Affiliation(s)
- Lia Furlaneto Marega
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Janine Schincariol Sabino
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Marcus Vinicius Pedroni
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Marcelo Teocchi
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Carolina Lanaro
- Hematology and Hemotherapy Center - Hemocentro Campinas, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | | | - Irene Pereira Dos Santos
- Flow Cytometry Laboratory, Hematology and Hemotherapy Center, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | - Fernando Ferreira Costa
- Hematology and Hemotherapy Center - Hemocentro Campinas, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | - Maria Marluce Dos Santos Vilela
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil.
| |
Collapse
|
48
|
Zhou XQ, Mao XM, Fan R, Li SY, Shang J, Zhang T, Li RH, Li HQ, Hui Y, Chen WH, Wang ZX, Shen DY. Trilobolide-6-O-isobutyrate suppresses hepatocellular carcinoma tumorigenesis through inhibition of IL-6/STAT3 signaling pathway. Phytother Res 2021; 35:5741-5753. [PMID: 34355433 DOI: 10.1002/ptr.7233] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/23/2021] [Accepted: 07/21/2021] [Indexed: 11/06/2022]
Abstract
Currently available therapies for hepatocellular carcinoma (HCC), with a high morbidity and high mortality, are only marginally effective and with sharp adverse side effects, which makes it compulsory to explore novel and more effective anticancer molecules. Chinese medicinal herbs exhibited prominent anticancer effects and were applied to supplement clinical cancer treatment. Here, we reported a compound, trilobolide-6-O-isobutyrate (TBB), isolated from the flowers of Wedelia trilobata with a markedly cytotoxic effect on HCC cells. We found that TBB time- and dose-dependently inhibited HCC cells' growth and colony formation in vitro. Moreover, TBB induced cell cycle arrest at the G2/M phase, mitochondrial caspase-dependent apoptosis, and suppressed migration and invasion, as well as the glycolysis of HCC cells. Mechanistically, our data indicated that TBB inhibited the STAT3 pathway activation by directly interacting with the TYR 640/657 sites of the STAT3 protein and decreasing the level of p-STAT3. TBB also regulated the expression of PCNA, Ki67, Cyclin B1, Cyclin E, Bax, Bcl2, MMP2/9, and PGK1 through the inhibition of the IL-6/STAT3 signaling pathway. Lastly, we confirmed that TBB effectively eliminated tumor growth without causing overt toxicity to healthy tissues in the xenograft tumor model. The exploration of anticancer activity and the underlying mechanism of TBB suggested its usage as a promising chemotherapeutic agent for HCC.
Collapse
Affiliation(s)
- Xiu-Qiao Zhou
- Key Laboratory of Tropical Medicinal Resources Chemistry of Ministry of Education, Hainan Normal University, Haikou, China
| | - Xiao-Mei Mao
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Rui Fan
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Si-Yang Li
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jin Shang
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Tong Zhang
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Rui-Han Li
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hui-Qi Li
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Hui
- Key Laboratory of Tropical Medicinal Resources Chemistry of Ministry of Education, Hainan Normal University, Haikou, China
| | - Wen-Hao Chen
- Key Laboratory of Tropical Medicinal Resources Chemistry of Ministry of Education, Hainan Normal University, Haikou, China
| | - Zhan-Xiang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Dong-Yan Shen
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
49
|
Shi J, Su Q, Han F, Chen W, Zhang D, Xu B. MiR-337 suppresses pancreatic cancer development via STAT3/Wnt/β-catenin axis. Anticancer Drugs 2021; 32:681-692. [PMID: 33587353 DOI: 10.1097/cad.0000000000001044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
MiRNA is an important regulator of tumorigenesis and tumor progression. MiR-337 expression was increased in pancreatic cancer tissues and it was associated with patients' survival. This study aimed to explore the influence and the potential working mechanism of miR-337 on the malignant behaviors of pancreatic cancer cells. MiR-337 expression was detected by qRT-PCR. The expression levels of STAT3, epithelial-mesenchymal transition-related genes and Wnt/β-Catenin pathway genes were evaluated by qRT-PCR and western blot. Cell counting kit -8 and colony formation assays were conducted to examine the proliferation of AsPC-1 and SW1990 cells. Wound healing and transwell assays were performed to determine the migration and invasion of AsPC-1 and SW1990 cells. The predicted target gene of miR-337 was verified by luciferase reporter assay. The expression of miR-337 was decreased and STAT3 expression was increased in pancreatic cancer tissues as well as tumor cells. Overexpression of miR-337 suppressed proliferation, invasion and migration of AsPC-1 and SW1990 cells. MiR-337 targeted 3'UTR of STAT3 and inhibited STAT3 expression. In addition, exogenous STAT3 partially restored the inhibitory role of miR-337 on proliferation, invasion and migration of AsPC-1 and SW1990 cells. Moreover, miR-337 impeded the expression of Wnt/β-catenin pathway-related genes. Through the saving experiment, we found that the inhibitory effect of miR-337 on AsPC-1 and SW1990 cells was abolished by the addition of LiCl. These outcomes expounded that miR-337 inactivated the Wnt/β-catenin pathway to suppress the malignant behaviors of pancreatic cancer cells through targeting STAT3. This study may provide a novel biomarker for diagnosis and a new therapeutic target for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Jin Shi
- Department of General Surgery, The 960th Hospital of the Chinese people's Liberation Army, Zhoucun District, Zibo
| | - Qingliang Su
- Department of General Surgery, Chengwu County People's Hospital, Chengwu County, Heze
| | | | | | - Donghua Zhang
- Department of Cancer Chemotherapy, Zhangqiu District People's Hospital, Zhangqiu District, Jinan
| | - Baoli Xu
- Department of General Surgery, Linyi People's Hospital, Lanshan District, Linyi, Shandong, China
| |
Collapse
|
50
|
Diallo M, Herrera F. The role of understudied post-translational modifications for the behavior and function of Signal Transducer and Activator of Transcription 3. FEBS J 2021; 289:6235-6255. [PMID: 34235865 DOI: 10.1111/febs.16116] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/16/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022]
Abstract
The Signal Transducer and Activator of Transcription (STAT) family of transcription factors is involved in inflammation, immunity, development, cancer, and response to injury, among other biological phenomena. Canonical STAT signaling is often represented as a 3-step pathway involving the sequential activation of a membrane receptor, an intermediate kinase, and a STAT transcription factor. The rate-limiting phosphorylation at a highly conserved C-terminal tyrosine residue determines the nuclear translocation and transcriptional activity of STATs. This apparent simplicity is actually misleading and can hardly explain the pleiotropic nature of STATs, the existence of various noncanonical STAT pathways, or the key role of the N-terminal domain in STAT functions. More than 80 post-translational modifications (PTMs) have been identified for STAT3, but their functions remain barely understood. Here, we provide a brief but comprehensive overview of these underexplored PTMs and their role on STAT3 canonical and noncanonical functions. A less tyrosine-centric point of view may be required to advance our understanding of STAT signaling.
Collapse
Affiliation(s)
- Mickael Diallo
- Faculdade de Ciências da Universidade de Lisboa, Cell Structure and Dynamics Laboratory, BioISI - Instituto de Biosistemas e Ciências integrativas, Lisbon, Portugal.,MOSTMICRO Research Unit, Instituto de Tecnologia Química e Biológica (ITQB-NOVA), Universidade Nova de Lisboa, Oeiras, Portugal
| | - Federico Herrera
- Faculdade de Ciências da Universidade de Lisboa, Cell Structure and Dynamics Laboratory, BioISI - Instituto de Biosistemas e Ciências integrativas, Lisbon, Portugal.,MOSTMICRO Research Unit, Instituto de Tecnologia Química e Biológica (ITQB-NOVA), Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|