1
|
Consonni F, Del Medico G, Stagi S, Gambineri E. Growth Failure in STAT3 Gain-of-Function Syndrome Persists After Hematopoietic Stem Cell Transplantation. J Clin Immunol 2024; 44:134. [PMID: 38787482 DOI: 10.1007/s10875-024-01727-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Affiliation(s)
- Filippo Consonni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Giulia Del Medico
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Stefano Stagi
- Department of Health Sciences, University of Florence, Florence, Italy
- Auxoendocrinology Division, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Eleonora Gambineri
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children's Hospital IRCCS, Florence, Italy.
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy.
| |
Collapse
|
2
|
Zhou Q, Chen D, Yu J, Zheng B, Zhou W, Jia Z, Zhang A, Gu W. A novel gain-of-function STAT3 variant in infantile-onset diabetes associated with multiorgan autoimmunity. Mol Genet Genomic Med 2024; 12:e2407. [PMID: 38404237 PMCID: PMC10895381 DOI: 10.1002/mgg3.2407] [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: 09/15/2023] [Revised: 01/27/2024] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Germline gain-of-function (GOF) variants in the signal transducer and activator of transcription 3 (STAT3) gene lead to a rare inherited disorder characterized by early-onset multiorgan autoimmunity. METHODS We described a Chinese patient with infantile-onset diabetes and multiorgan autoimmunity. The patient presented with early-onset type 1 diabetes and autoimmune hypothyroidism at 7 months. During the 7.5-year follow-up, she developed pseudo-celiac enteropathy at 1 year of age and showed severe growth retardation. Whole-exome sequencing was performed and the novel variant was further assessed by in vitro functional assays. RESULTS Whole-exome sequencing revealed a novel variant (c.1069G>A, p.Glu357Lys) in the DNA-binding domain of STAT3. In vitro functional studies revealed that p.Glu357Lys was a GOF variant by increasing STAT3 transcriptional activity and phosphorylation. In addition, the STAT3 Glu357Lys variant caused dysregulation of insulin gene expression by enhancing transcriptional inhibition of the insulin gene enhancer binding protein factor 1 (ISL1). CONCLUSION In the current study, we describe clinical manifestations and identify a novel STAT3 GOF variant (c.1069G>A) in a Chinese patient. This activating variant impairs insulin expression by increasing transcriptional inhibition of its downstream transcription factor ISL1, which could be involved in the pathogenesis of early-onset diabetes.
Collapse
Affiliation(s)
- Qiaoli Zhou
- Department of EndocrinologyChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Dandan Chen
- Department of Child HealthcareLianyungang Maternal and Children's HospitalLianyungangChina
| | - Jing Yu
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Bixia Zheng
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Wei Zhou
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhanjun Jia
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Aihua Zhang
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Wei Gu
- Department of EndocrinologyChildren's Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
3
|
Alam A, Dhoundiyal S, Ahmad N, Rao GSNK. Unveiling Diabetes: Categories, Genetics, Diagnostics, Treatments, and Future Horizons. Curr Diabetes Rev 2024; 20:e180823219972. [PMID: 37594107 DOI: 10.2174/1573399820666230818092958] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/15/2023] [Accepted: 07/06/2023] [Indexed: 08/19/2023]
Abstract
Diabetes mellitus is a global epidemic affecting millions of individuals worldwide. This comprehensive review aims to provide a thorough understanding of the categorization, disease identity, genetic architecture, diagnosis, and treatment of diabetes. The categorization of diabetes is discussed, with a focus on type 1 and type 2 diabetes, as well as the lesser-known types, type 3 and type 4 diabetes. The geographical variation, age, gender, and ethnic differences in the prevalence of type 1 and type 2 diabetes are explored. The impact of disease identity on disease management and the role of autoimmunity in diabetes are examined. The genetic architecture of diabetes, including the interplay between genotype and phenotype, is discussed to enhance our understanding of the underlying mechanisms. The importance of insulin injection sites and the insulin signalling pathway in diabetes management are highlighted. The diagnostic techniques for diabetes are reviewed, along with advancements for improved differentiation between types. Treatment and management approaches, including medications used in diabetes management are presented. Finally, future perspectives are discussed, emphasizing the need for further research and interventions to address the global burden of diabetes. This review serves as a valuable resource for healthcare professionals, researchers, and policymakers, providing insights to develop targeted strategies for the prevention, diagnosis, and management of this complex disease.
Collapse
Affiliation(s)
- Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shivang Dhoundiyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Niyaz Ahmad
- Department of Pharmaceutical Analysis, Green Research Lab, Green Industrial Company, Second Industrial Area, Riyadh 14334, Saudi Arabia
| | - G S N Koteswara Rao
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| |
Collapse
|
4
|
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: 22] [Impact Index Per Article: 22.0] [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
|
5
|
Toni L, Plachy L, Dusatkova P, Amaratunga SA, Elblova L, Sumnik Z, Kolouskova S, Snajderova M, Obermannova B, Pruhova S, Lebl J. The Genetic Landscape of Children Born Small for Gestational Age with Persistent Short Stature. Horm Res Paediatr 2023; 97:40-52. [PMID: 37019085 DOI: 10.1159/000530521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
INTRODUCTION Among children born small for gestational age, 10-15% fail to catch up and remain short (SGA-SS). The underlying mechanisms are mostly unknown. We aimed to decipher genetic aetiologies of SGA-SS within a large single-centre cohort. METHODS Out of 820 patients treated with growth hormone (GH), 256 were classified as SGA-SS (birth length and/or birth weight <-2 SD for gestational age and life-minimum height <-2.5 SD). Those with the DNA triplet available (child and both parents) were included in the study (176/256). Targeted testing (karyotype/FISH/MLPA/specific Sanger sequencing) was performed if a specific genetic disorder was clinically suggestive. All remaining patients underwent MS-MLPA to identify Silver-Russell syndrome, and those with unknown genetic aetiology were subsequently examined using whole-exome sequencing or targeted panel of 398 growth-related genes. Genetic variants were classified using ACMG guidelines. RESULTS The genetic aetiology was elucidated in 74/176 (42%) children. Of these, 12/74 (16%) had pathogenic or likely pathogenic (P/LP) gene variants affecting pituitary development (LHX4, OTX2, PROKR2, PTCH1, POU1F1), the GH-IGF-1 or IGF-2 axis (GHSR, IGFALS, IGF1R, STAT3, HMGA2), 2/74 (3%) the thyroid axis (TRHR, THRA), 17/74 (23%) the cartilaginous matrix (ACAN, various collagens, FLNB, MATN3), and 7/74 (9%) the paracrine chondrocyte regulation (FGFR3, FGFR2, NPR2). In 12/74 (16%), we revealed P/LP affecting fundamental intracellular/intranuclear processes (CDC42, KMT2D, LMNA, NSD1, PTPN11, SRCAP, SON, SOS1, SOX9, TLK2). SHOX deficiency was found in 7/74 (9%), Silver-Russell syndrome in 12/74 (16%) (11p15, UPD7), and miscellaneous chromosomal aberrations in 5/74 (7%) children. CONCLUSIONS The high diagnostic yield sheds a new light on the genetic landscape of SGA-SS, with a central role for the growth plate with substantial contributions from the GH-IGF-1 and thyroid axes and intracellular regulation and signalling.
Collapse
Affiliation(s)
- Ledjona Toni
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Lukas Plachy
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Petra Dusatkova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Shenali Anne Amaratunga
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Lenka Elblova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Zdenek Sumnik
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Stanislava Kolouskova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Marta Snajderova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Barbora Obermannova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Stepanka Pruhova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Jan Lebl
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| |
Collapse
|
6
|
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, 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] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [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
|
7
|
Mulvihill C, Fitzgerald K, O'Regan E, Healy CM, Leahy TR. Successful treatment of oral potentially malignant lesions with ruxolitinib in STAT3 gain-of-function disease. Oral Oncol 2022; 132:106015. [PMID: 35809509 DOI: 10.1016/j.oraloncology.2022.106015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 11/16/2022]
Affiliation(s)
- Ciara Mulvihill
- Department of Oral and Maxillofacial Surgery, Oral Medicine and Oral Pathology, Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland.
| | - Kirsten Fitzgerald
- Departments of Paediatric Dentistry, Children's Health Ireland at Crumlin, Dublin and Dublin Dental University Hospital, Dublin, Ireland
| | - Esther O'Regan
- Department of Oral and Maxillofacial Surgery, Oral Medicine and Oral Pathology, Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Claire M Healy
- Department of Oral and Maxillofacial Surgery, Oral Medicine and Oral Pathology, Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Timothy Ronan Leahy
- Department of Paediatric Immunology and Infectious Diseases, Children's Health Ireland at Crumlin, Dublin, Ireland; Department of Paediatrics, School of Medicine, University of Dublin, Trinity College, Dublin, Ireland
| |
Collapse
|
8
|
Schiavo E, Martini B, Attardi E, Consonni F, Ciullini Mannurita S, Coniglio ML, Tellini M, Chiocca E, Fotzi I, Luti L, D'Alba I, Veltroni M, Favre C, Gambineri E. Autoimmune Cytopenias and Dysregulated Immunophenotype Act as Warning Signs of Inborn Errors of Immunity: Results From a Prospective Study. Front Immunol 2022; 12:790455. [PMID: 35058929 PMCID: PMC8765341 DOI: 10.3389/fimmu.2021.790455] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022] Open
Abstract
Inborn errors of immunity (IEI) are genetic disorders characterized by a wide spectrum of clinical manifestations, ranging from increased susceptibility to infections to significant immune dysregulation. Among these, primary immune regulatory disorders (PIRDs) are mainly presenting with autoimmune manifestations, and autoimmune cytopenias (AICs) can be the first clinical sign. Significantly, AICs in patients with IEI often fail to respond to first-line therapy. In pediatric patients, autoimmune cytopenias can be red flags for IEI. However, for these cases precise indicators or parameters useful to suspect and screen for a hidden congenital immune defect are lacking. Therefore, we focused on chronic/refractory AIC patients to perform an extensive clinical evaluation and multiparametric flow cytometry analysis to select patients in whom PIRD was strongly suspected as candidates for genetic analysis. Key IEI-associated alterations causative of STAT3 GOF disease, IKAROS haploinsufficiency, activated PI3Kδ syndrome (APDS), Kabuki syndrome and autoimmune lymphoproliferative syndrome (ALPS) were identified. In this scenario, a dysregulated immunophenotype acted as a potential screening tool for an early IEI diagnosis, pivotal for appropriate clinical management and for the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Ebe Schiavo
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Beatrice Martini
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Enrico Attardi
- Division of Hematology, Careggi University Hospital, Florence, Italy
| | - Filippo Consonni
- Meyer University Children's Hospital, University of Florence, Florence, Italy
| | - Sara Ciullini Mannurita
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Maria Luisa Coniglio
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Marco Tellini
- Meyer University Children's Hospital, University of Florence, Florence, Italy
| | - Elena Chiocca
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Ilaria Fotzi
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Laura Luti
- Division of Pediatric Oncology/Hematology, University Hospital of Pisa, Pisa, Italy
| | - Irene D'Alba
- Division of Pediatric Oncology/Hematology, University Hospital of Ospedali Riuniti, Ancona, Italy
| | - Marinella Veltroni
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Claudio Favre
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Eleonora Gambineri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy.,Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| |
Collapse
|
9
|
Vogel TP, Leiding JW, Cooper MA, Forbes Satter LR. STAT3 gain-of-function syndrome. Front Pediatr 2022; 10:770077. [PMID: 36843887 PMCID: PMC9948021 DOI: 10.3389/fped.2022.770077] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/28/2022] [Indexed: 02/11/2023] Open
Abstract
STAT3 gain-of-function (GOF) syndrome is a multi-organ primary immune regulatory disorder characterized by early onset autoimmunity. Patients present early in life, most commonly with lymphoproliferation, autoimmune cytopenias, and growth delay. However, disease is often progressive and can encompass a wide range of clinical manifestations such as: enteropathy, skin disease, pulmonary disease, endocrinopathy, arthritis, autoimmune hepatitis, and rarely neurologic disease, vasculopathy, and malignancy. Treatment of the autoimmune and immune dysregulatory features of STAT3-GOF patients relies heavily on immunosuppression and is often challenging and fraught with complications including severe infections. Defects in the T cell compartment leading to effector T cell accumulation and decreased T regulatory cells may contribute to autoimmunity. While T cell exhaustion and apoptosis defects likely contribute to the lymphoproliferative phenotype, no conclusive correlations are yet established. Here we review the known mechanistic and clinical characteristics of this heterogenous PIRD.
Collapse
Affiliation(s)
- Tiphanie P Vogel
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX, United States
| | - Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Baltimore, MD, United States.,Orlando Health Arnold Palmer Hospital for Children, Orlando, FL, United States
| | - Megan A Cooper
- Division of Rheumatology and Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Lisa R Forbes Satter
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
10
|
Korenfeld D, Roussak K, Dinkel S, Vogel TP, Pollack H, Levy J, Leiding JW, Milner J, Cooper M, Klechevsky E. STAT3 Gain-of-Function Mutations Underlie Deficiency in Human Nonclassical CD16 + Monocytes and CD141 + Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:2423-2432. [PMID: 34654687 DOI: 10.4049/jimmunol.2000841] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/14/2021] [Indexed: 12/24/2022]
Abstract
Genetic analysis of human inborn errors of immunity has defined the contribution of specific cell populations and molecular pathways in the host defense against infection. The STAT family of transcription factors orchestrate hematopoietic cell differentiation. Patients with de novo activating mutations of STAT3 present with multiorgan autoimmunity, lymphoproliferation, and recurrent infections. We conducted a detailed characterization of the blood monocyte and dendritic cell (DC) subsets in patients with gain-of-function (GOF) mutations across the gene. We found a selective deficiency in circulating nonclassical CD16+ and intermediate CD16+CD14+ monocytes and a significant increase in the percentage of classical CD14+ monocytes. This suggests a role for STAT3 in the transition of classical CD14+ monocytes into the CD16+ nonclassical subset. Developmentally, ex vivo-isolated STAT3GOF CD14+ monocytes fail to differentiate into CD1a+ monocyte-derived DCs. Moreover, patients with STAT3GOF mutations display reduced circulating CD34+ hematopoietic progenitors and frequency of myeloid DCs. Specifically, we observed a reduction in the CD141+ DC population, with no difference in the frequencies of CD1c+ and plasmacytoid DCs. CD34+ hematopoietic progenitor cells from patients were found to differentiate into CD1c+ DCs, but failed to differentiate into CD141+ DCs indicating an intrinsic role for STAT3 in this process. STAT3GOF-differentiated DCs produced lower amounts of CCL22 than healthy DCs, which could further explain some of the patient pathological phenotypes. Thus, our findings provide evidence that, in humans, STAT3 serves to regulate development and differentiation of nonclassical CD16+ monocytes and a subset of myeloid DCs.
Collapse
Affiliation(s)
- Daniel Korenfeld
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Kate Roussak
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Sabrina Dinkel
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Tiphanie P Vogel
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University School of Medicine, St. Louis, MO
| | - Henry Pollack
- Department of Pediatrics, New York University School of Medicine, New York, NY
| | - Joseph Levy
- Department of Pediatrics, New York University School of Medicine, New York, NY
| | - Jennifer W Leiding
- Department of Pediatrics, Division of Allergy and Immunology, University of South Florida, Tampa, FL; and
| | - Joshua Milner
- Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Megan Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University School of Medicine, St. Louis, MO
| | - Eynav Klechevsky
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO;
| |
Collapse
|
11
|
Warshauer JT, Belk JA, Chan AY, Wang J, Gupta AR, Shi Q, Skartsis N, Peng Y, Phipps JD, Acenas D, Smith JA, Tamaki SJ, Tang Q, Gardner JM, Satpathy AT, Anderson MS. A human mutation in STAT3 promotes type 1 diabetes through a defect in CD8+ T cell tolerance. J Exp Med 2021; 218:212280. [PMID: 34115115 PMCID: PMC8203485 DOI: 10.1084/jem.20210759] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 12/16/2022] Open
Abstract
Naturally occurring cases of monogenic type 1 diabetes (T1D) help establish direct mechanisms driving this complex autoimmune disease. A recently identified de novo germline gain-of-function (GOF) mutation in the transcriptional regulator STAT3 was found to cause neonatal T1D. We engineered a novel knock-in mouse incorporating this highly diabetogenic human STAT3 mutation (K392R) and found that these mice recapitulated the human autoimmune diabetes phenotype. Paired single-cell TCR and RNA sequencing revealed that STAT3-GOF drives proliferation and clonal expansion of effector CD8+ cells that resist terminal exhaustion. Single-cell ATAC-seq showed that these effector T cells are epigenetically distinct and have differential chromatin architecture induced by STAT3-GOF. Analysis of islet TCR clonotypes revealed a CD8+ cell reacting against known antigen IGRP, and STAT3-GOF in an IGRP-reactive TCR transgenic model demonstrated that STAT3-GOF intrinsic to CD8+ cells is sufficient to accelerate diabetes onset. Altogether, these findings reveal a diabetogenic CD8+ T cell response that is restrained in the presence of normal STAT3 activity and drives diabetes pathogenesis.
Collapse
Affiliation(s)
- Jeremy T. Warshauer
- Diabetes Center, University of California, San Francisco, San Francisco, CA,Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Julia A. Belk
- Department of Computer Science, Stanford University, Stanford, CA
| | - Alice Y. Chan
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Jiaxi Wang
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Alexander R. Gupta
- Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Quanming Shi
- Department of Pathology, Stanford University, Stanford, CA
| | - Nikolaos Skartsis
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Yani Peng
- Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Jonah D. Phipps
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Dante Acenas
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Jennifer A. Smith
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Stanley J. Tamaki
- Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - James M. Gardner
- Diabetes Center, University of California, San Francisco, San Francisco, CA,Department of Surgery, University of California, San Francisco, San Francisco, CA
| | | | - Mark S. Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, CA,Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA,Department of Medicine, University of California, San Francisco, San Francisco, CA,Correspondence to Mark S. Anderson:
| |
Collapse
|
12
|
[Immune dysregulation syndrome caused by STAT3 gene mutation: a complicated case study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021. [PMID: 33840413 PMCID: PMC8050543 DOI: 10.7499/j.issn.1008-8830.2012167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A boy, aged 4 years and 6 months, had disease onset of fever, cough, pale complexion, and weakness, with hepatosplenomegaly, lymphadenectasis, and pancytopenia. He had been having repeated respiratory and digestive tract infections. Gene detection showed a pathogenic heterozygous mutation, c.C2147 > T(p.T716M), in the STAT3 gene. The boy was thus diagnosed with immune dysregulation syndrome. Anti-infective therapy and irregular corticosteroid therapy had an unsatisfactory effect in the early stage, but the symptoms improved after regular corticosteroid therapy. This article reported the case of immune dysregulation syndrome caused by STAT3 gene mutation and summarized the epidemiology, clinical features, diagnosis, and treatment of this disease, which can provide a reference for early diagnosis, treatment, and future studies of this disease.
Collapse
|
13
|
Faletti L, Ehl S, Heeg M. Germline STAT3 gain-of-function mutations in primary immunodeficiency: Impact on the cellular and clinical phenotype. Biomed J 2021; 44:412-421. [PMID: 34366294 PMCID: PMC8514798 DOI: 10.1016/j.bj.2021.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 12/25/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a key transcription factor involved in regulation of immune cell activation and differentiation. Recent discoveries highlight the role of germline activating STAT3 mutations in inborn errors of immunity characterized by early-onset multi-organ autoimmunity and lymphoproliferation. Much progress has been made in defining the clinical spectrum of STAT3 GOF disease and unraveling the molecular and cellular mechanisms underlying this disease. In this review, we summarize our current understanding of the disease and discuss the clinical phenotype, diagnostic approach, cellular and molecular effects of STAT3 GOF mutations and therapeutic concepts for these patients.
Collapse
Affiliation(s)
- Laura Faletti
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
14
|
Wang W, Liu L, Hui X, Wang Y, Ying W, Zhou Q, Hou J, Yang M, Sun B, Sun J, Wang X. Efficacy of tocilizumab therapy in a patient with severe pancytopenia associated with a STAT3 gain-of-function mutation. BMC Immunol 2021; 22:19. [PMID: 33731004 PMCID: PMC7968248 DOI: 10.1186/s12865-021-00411-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/02/2021] [Indexed: 11/20/2022] Open
Abstract
Background We aimed to report the clinical characteristics, immunological features, and treatment of one patient with a de novo STAT3 gain-of-function mutation identified by next generation sequencing. We investigated the efficacy of tocilizumab therapy in immune dysregulation diseases caused by STAT3 mutation. Results The patient was a 16-year-old girl. She presented with recurrent respiratory infections and chronic diarrhea after birth. She had life-threatening autoimmune pancytopenia at 14 years old. After receiving glucocorticoid therapy, she developed diabetes. However, her pancytopenia relapsed when the glucocorticoid was tapered. Next-generation sequencing showed a de novo heterozygous mutation in the STAT3 gene, c.1261G > A (p. G421R), which was previously described as a gain-of-function mutation. After tocilizumab therapy, her pancytopenia fully resolved, and insulin and glucocorticoid therapies were gradually discontinued within 12 months. She had lymphopenia and an inverted CD4/CD8 ratio before therapy. Lymphocyte subpopulation analysis indicated an expansion of effector memory CD4+, effector memory CD8+ and central memory CD4+ T cells. The proportions of memory B cells and naive CD4+ T cells were decreased, and the proportion of naïve B cells was increased. None of the abnormal lymphocytic changes improved significantly. STAT3 GOF mutations were identified by next gene sequencing in those with early-onset multi-organ autoimmunity. Including our patient, 13 patients with STAT3 GOF mutations received targeted treatment. Twelve of them were treated with tocilizumab alone or combination tocilizumab with JAK inhibitor, and ten patients improved. Conclusions Gene sequencing should be performed for patients with early-onset refractory or multiorgan immune dysregulation diseases. Targeted drugs can effectively improve the clinical problems associated with STAT3 gain-of-function mutations, while nontargeted immunosuppressive therapy is usually insufficient. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00411-1.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Luyao Liu
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Xiaoying Hui
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Ying Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Wenjing Ying
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Qinhua Zhou
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Jia Hou
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Mi Yang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Bijun Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Jinqiao Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
| | - Xiaochuan Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
| |
Collapse
|
15
|
Ciullini Mannurita S, Goda R, Schiavo E, Coniglio ML, Azzali A, Fotzi I, Tondo A, Tintori V, Frenos S, Sanvito MC, Vignoli M, Luceri C, Bigagli E, Grassi A, D'Elios MM, Favre C, Gambineri E. Case Report: Signal Transducer and Activator of Transcription 3 Gain-of-Function and Spectrin Deficiency: A Life-Threatening Case of Severe Hemolytic Anemia. Front Immunol 2021; 11:620046. [PMID: 33519826 PMCID: PMC7843414 DOI: 10.3389/fimmu.2020.620046] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/20/2020] [Indexed: 12/14/2022] Open
Abstract
STAT3 gain-of-function (GOF) mutations can be responsible for an incomplete phenotype mainly characterized by hematological autoimmunity, even in the absence of other organ autoimmunity, growth impairment, or severe infections. We hereby report a case with an incomplete form of STAT3 GOF intensified by a concomitant hereditary hematological disease, which misleads the diagnosis. The patient presented with lymphadenopathy, splenomegaly, hypogammaglobulinemia, and severe autoimmune hemolytic anemia (AIHA) with critical complications, including stroke. A Primary Immune Regulatory Disorders (PIRD) was suspected, and molecular analysis revealed a de novo STAT3 gain-of-function mutation. The response to multiple immune suppressive treatments was ineffective, and further investigations revealed a spectrin deficiency. Ultimately, hematopoietic stem cell transplantation from a matched unrelated donor was able to cure the patient. Our case shows an atypical presentation of STAT3 GOF associated with hereditary spherocytosis, and how achievement of a good long-term outcome depends on a strict clinical and laboratory monitoring, as well as on prompt therapeutic intervention.
Collapse
Affiliation(s)
- Sara Ciullini Mannurita
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Rayan Goda
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Ebe Schiavo
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Maria Luisa Coniglio
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Annachiara Azzali
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Ilaria Fotzi
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Annalisa Tondo
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Veronica Tintori
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Stefano Frenos
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Maria Chiara Sanvito
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Marina Vignoli
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Cristina Luceri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Elisabetta Bigagli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Alessia Grassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Claudio Favre
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Eleonora Gambineri
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
16
|
Gutiérrez M. Activating mutations of STAT3: Impact on human growth. Mol Cell Endocrinol 2020; 518:110979. [PMID: 32818584 DOI: 10.1016/j.mce.2020.110979] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023]
Abstract
The signal transducer and activator of transcription (STAT) 3 is the most ubiquitous member of the STAT family and fulfills fundamental functions in immune and non-immune cells. Mutations in the STAT3 gene lead to different human diseases. Germline STAT3 activating or gain-of-function (GOF) mutations result in early-onset multiorgan autoimmunity, lymphoproliferation, recurrent infections and short stature. Since the first description of the disease, the clinical manifestations of STAT3 GOF mutations have expanded considerably. However, due to the complexity of immunological characteristics in patients carrying STAT3 GOF mutations, most of attention was focused on the immune alterations. This review summarizes current knowledge on STAT3 GOF mutations with special focus on the growth defects, since short stature is a predominant feature in this condition. Underlying mechanisms of STAT3 GOF disease are still poorly understood, and potential effects of STAT3 GOF mutations on the growth hormone signaling pathway are unclear. Functional studies of STAT3 GOF mutations and the broadening of clinical growth-related data in these patients are necessary to better delineate implications of STAT3 GOF mutations on growth.
Collapse
Affiliation(s)
- Mariana Gutiérrez
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños R. Gutiérrez, Gallo 1360, Buenos Aires, CP1425EFD, Argentina.
| |
Collapse
|
17
|
Mauracher AA, Eekels JJM, Woytschak J, van Drogen A, Bosch A, Prader S, Felber M, Heeg M, Opitz L, Trück J, Schroeder S, Adank E, Klocperk A, Haralambieva E, Zimmermann D, Tantou S, Kotsonis K, Stergiou A, Kanariou MG, Ehl S, Boyman O, Sediva A, Renella R, Schmugge M, Vavassori S, Pachlopnik Schmid J. Erythropoiesis defect observed in STAT3 GOF patients with severe anemia. J Allergy Clin Immunol 2019; 145:1297-1301. [PMID: 31857100 DOI: 10.1016/j.jaci.2019.11.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 01/29/2023]
Affiliation(s)
- Andrea A Mauracher
- Division of Immunology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Julia J M Eekels
- Division of Haematology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Janine Woytschak
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Audrey van Drogen
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Alessandra Bosch
- Division of Haematology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Seraina Prader
- Division of Immunology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Matthias Felber
- Division of Immunology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland; Division of Haematology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Maximillian Heeg
- Institute of Immunodeficiency and Center for Chronic Immunodeficiency, University Medical Center, Freiburg, Germany
| | - Lennart Opitz
- Division of Immunology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland; Functional Genomics Center Zürich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Johannes Trück
- Division of Immunology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland; University of Zurich, Zurich, Switzerland
| | - Silke Schroeder
- Division of Rheumatology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Eva Adank
- Division of Rheumatology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Adam Klocperk
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Eugenia Haralambieva
- Department of Pathology University and University Hospital Zurich, Zurich, Switzerland
| | - Dieter Zimmermann
- Department of Pathology University and University Hospital Zurich, Zurich, Switzerland
| | - Sofia Tantou
- First Department of Paediatrics, Aghia Sophia Children's Hospital, Athens, Greece
| | - Kosmas Kotsonis
- First Department of Paediatrics, Aghia Sophia Children's Hospital, Athens, Greece
| | - Aikaterini Stergiou
- First Department of Paediatrics, Aghia Sophia Children's Hospital, Athens, Greece
| | - Maria G Kanariou
- First Department of Paediatrics, Aghia Sophia Children's Hospital, Athens, Greece
| | - Stephan Ehl
- Institute of Immunodeficiency and Center for Chronic Immunodeficiency, University Medical Center, Freiburg, Germany
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anna Sediva
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Raffaele Renella
- Pediatric Hematology-Oncology Unit, Division of Pediatrics, Lausanne University Hospital, Lausanne, Switzerland
| | - Markus Schmugge
- Division of Haematology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Stefano Vavassori
- Division of Immunology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Jana Pachlopnik Schmid
- Division of Immunology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland; University of Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Jägle S, Heeg M, Grün S, Rensing-Ehl A, Maccari ME, Klemann C, Jones N, Lehmberg K, Bettoni C, Warnatz K, Grimbacher B, Biebl A, Schauer U, Hague R, Neth O, Mauracher A, Pachlopnik Schmid J, Fabre A, Kostyuchenko L, Führer M, Lorenz MR, Schwarz K, Rohr J, Ehl S. Distinct molecular response patterns of activating STAT3 mutations associate with penetrance of lymphoproliferation and autoimmunity. Clin Immunol 2019; 210:108316. [PMID: 31770611 DOI: 10.1016/j.clim.2019.108316] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/16/2019] [Accepted: 11/20/2019] [Indexed: 01/12/2023]
Abstract
Germline STAT3 gain-of-function (GOF) mutations have been linked to poly-autoimmunity and lymphoproliferation with variable expressivity and incomplete penetrance. Here we studied the impact of 17 different STAT3 GOF mutations on the canonical STAT3 signaling pathway and correlated the molecular results with clinical manifestations. The mutations clustered in three groups. Group 1 mutants showed altered STAT3 phosphorylation kinetics and strong basal transcriptional activity. They were associated with the highest penetrance of lymphoproliferation and autoimmunity. Group 2 mutants showed a strongly inducible transcriptional reporter activity and were clinically less penetrant. Group 3 mutants were mostly located in the DNA binding domain and showed the strongest DNA binding affinity despite a poor transcriptional reporter response. Thus, the GOF effect of STAT3 mutations is determined by a heterogeneous response pattern at the molecular level. The correlation of response pattern and clinical penetrance indicates a significant contribution of mutation-determined effects on disease manifestations.
Collapse
Affiliation(s)
- Sabine Jägle
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Pediatrics, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sarah Grün
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anne Rensing-Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria Elena Maccari
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Pediatrics, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Klemann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| | - Neil Jones
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Kai Lehmberg
- Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Claudia Bettoni
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| | - Klaus Warnatz
- Divivion Immunodeficiency (CCI), Department of Rheumatology and Clinical Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; DZIF, German Center for Infection Research, Satellite Center, Freiburg, Germany; Resist - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Ariane Biebl
- Department of Paediatric and Adolescent Medicine, Kepler University Hospital Linz, Linz, Austria
| | - Uwe Schauer
- University Children's Hospital, Ruhr University Bochum, Bochum, Germany
| | - Rosie Hague
- Paediatric Infectious Diseases and Immunology, Royal Hospital for Children, Glasgow, UK
| | - Olaf Neth
- Paediatric Infectious Diseases, Rheumatology and Immunology, Hospital Universitario Virgen del Rocío, Instituto de Bioinvestigacion (IBIS), Sevilla, Spain
| | - Andrea Mauracher
- Division of Immunology, University Children's Hospital Zurich, Children's Research Center (CRC), Zurich, Switzerland
| | - Jana Pachlopnik Schmid
- Division of Immunology, University Children's Hospital Zurich, Children's Research Center (CRC), Zurich, Switzerland
| | - Alexandre Fabre
- Service de Pédiatrie Multidisciplinaire, Hôpital de la Timone, APHM, Marseille, France; Aix Marseille Univ, INSERM, MMG, Marseille, France
| | - Larysa Kostyuchenko
- Center of Pediatric Immunology, Western Ukrainian Specialized Children's Medical Centre, Lviv, Ukraine
| | - Marita Führer
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | | | - Klaus Schwarz
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany; Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg, Hessen, Ulm, Germany
| | - Jan Rohr
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Pediatrics, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Pediatrics, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
19
|
Vasques GA, Andrade NLM, Correa FA, Jorge AAL. Update on new GH-IGF axis genetic defects. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:608-617. [PMID: 31939486 PMCID: PMC10522240 DOI: 10.20945/2359-3997000000191] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/19/2019] [Indexed: 11/23/2022]
Abstract
The somatotropic axis is the main hormonal regulator of growth. Growth hormone (GH), also known as somatotropin, and insulin-like growth factor 1 (IGF-1) are the key components of the somatotropic axis. This axis has been studied for a long time and the knowledge of how some molecules could promote or impair hormones production and action has been growing over the last decade. The enhancement of large-scale sequencing techniques has expanded the spectrum of known genes and several other candidate genes that could affect the GH-IGF1-bone pathway. To date, defects in more than forty genes were associated with an impairment of the somatotropic axis. These defects can affect from the secretion of GH to the bioavailability and action of IGF-1. Affected patients present a large heterogeneous group of conditions associated with growth retardation. In this review, we focus on the description of the GH-IGF axis genetic defects reported in the last decade. Arch Endocrinol Metab. 2019;63(6):608-17.
Collapse
Affiliation(s)
- Gabriela A. Vasques
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrasil Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular (LIM25), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrasil Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Nathalia L. M. Andrade
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrasil Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular (LIM25), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrasil Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Fernanda A. Correa
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrasil Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Alexander A. L. Jorge
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrasil Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular (LIM25), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrasil Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
20
|
Strakova V, Elblova L, Johnson MB, Dusatkova P, Obermannova B, Petruzelkova L, Kolouskova S, Snajderova M, Fronkova E, Svaton M, Lebl J, Hattersley AT, Sumnik Z, Pruhova S. Screening of monogenic autoimmune diabetes among children with type 1 diabetes and multiple autoimmune diseases: is it worth doing? J Pediatr Endocrinol Metab 2019; 32:1147-1153. [PMID: 31483759 DOI: 10.1515/jpem-2019-0261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 08/09/2019] [Indexed: 11/15/2022]
Abstract
Background Paediatric type 1 diabetes (T1D) and rare syndromes of monogenic multi-organ autoimmunity share basic features such as full insulin dependency and the presence of circulating beta-cell autoantibodies. However, the aetiopathogenesis, natural course and treatment of these conditions differ; therefore, monogenic multi-organ autoimmunity requires early recognition. We aimed to search for these monogenic conditions among a large cohort of children with T1D. Methods Of 519 children with T1D followed-up in a single centre, 18 had multiple additional autoimmune conditions - either autoimmune thyroid disease (AITD) and coeliac disease (CD) or at least one additional organ-specific autoimmune condition in addition to AITD or CD. These 18 children were tested by direct Sanger sequencing (four patients with a suggestive phenotype of immune dysregulation, polyendocrinopathy, enteropathy, X-linked [IPEX] or signal transducer and activator of transcription 3 [STAT3]- and cytotoxic T-lymphocyte protein 4 [CTLA4]-associated syndromes) or by whole-exome sequencing (WES) focused on autoimmune regulator (AIRE), forkhead box protein 3 (FOXP3), CTLA4, STAT3, signal transducer and activator of transcription 1 (STAT1), lipopolysaccharide-responsive and beige-like anchor protein (LRBA) and interleukin-2 receptor subunit α (IL2RA) genes. In addition, we assessed their T1D genetic risk score (T1D-GRS). Results We identified novel variants in FOXP3, STAT3 and CTLA4 in four cases. All patients had a severe phenotype suggestive of a single gene defect. No variants were identified in the remaining 14 patients. T1D-GRS varied among the entire cohort; four patients had scores below the 25th centile including two genetically confirmed cases. Conclusions A monogenic cause of autoimmune diabetes was confirmed only in four patients. Genetic screening for monogenic autoimmunity in children with a milder phenotype and a combination of AITD and CD is unlikely to identify a monogenic cause. In addition, the T1D-GRS varied among individual T1D patients.
Collapse
Affiliation(s)
- Veronika Strakova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Lenka Elblova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Matthew B Johnson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Petra Dusatkova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Barbora Obermannova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Lenka Petruzelkova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Stanislava Kolouskova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Marta Snajderova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Eva Fronkova
- Department of Paediatric Haematology and Oncology, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Michael Svaton
- Department of Paediatric Haematology and Oncology, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Jan Lebl
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Zdenek Sumnik
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Stepanka Pruhova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
21
|
Yi R, Yang L, Zeng S, Su Y. Different expression profile of mRNA and long noncoding RNA in autoimmune thyroid diseases patients. J Cell Biochem 2019; 120:19442-19456. [PMID: 31452253 DOI: 10.1002/jcb.29233] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/23/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Ruhai Yi
- Endocrinology Department The First Affiliated Hospital of Fujian Medical University, Diabetes Research Insititute of Fujian Province Fuzhou Fujian Province People's Republic of China
| | - Liyong Yang
- Endocrinology Department The First Affiliated Hospital of Fujian Medical University, Diabetes Research Insititute of Fujian Province Fuzhou Fujian Province People's Republic of China
| | - Saifan Zeng
- Pathology Department The First Affiliated Hospital of Fujian Medical University China
| | - Yueqing Su
- Center of Neonatal Screening, Fujian Provincal Maternity and Children's Hospital Affiliated Hospital of Fujian Medical University Fuzhou Fujian Province China
| |
Collapse
|
22
|
Storr HL, Chatterjee S, Metherell LA, Foley C, Rosenfeld RG, Backeljauw PF, Dauber A, Savage MO, Hwa V. Nonclassical GH Insensitivity: Characterization of Mild Abnormalities of GH Action. Endocr Rev 2019; 40:476-505. [PMID: 30265312 PMCID: PMC6607971 DOI: 10.1210/er.2018-00146] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022]
Abstract
GH insensitivity (GHI) presents in childhood with growth failure and in its severe form is associated with extreme short stature and dysmorphic and metabolic abnormalities. In recent years, the clinical, biochemical, and genetic characteristics of GHI and other overlapping short stature syndromes have rapidly expanded. This can be attributed to advancing genetic techniques and a greater awareness of this group of disorders. We review this important spectrum of defects, which present with phenotypes at the milder end of the GHI continuum. We discuss their clinical, biochemical, and genetic characteristics. The objective of this review is to clarify the definition, identification, and investigation of this clinically relevant group of growth defects. We also review the therapeutic challenges of mild GHI.
Collapse
Affiliation(s)
- Helen L Storr
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Sumana Chatterjee
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Louise A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Corinne Foley
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ron G Rosenfeld
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon
| | - Philippe F Backeljauw
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Andrew Dauber
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Martin O Savage
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Vivian Hwa
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
23
|
Goudouris ES, Segundo GRS, Poli C. Repercussions of inborn errors of immunity on growth. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2019. [DOI: 10.1016/j.jpedp.2019.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
24
|
Goudouris ES, Segundo GRS, Poli C. Repercussions of inborn errors of immunity on growth. J Pediatr (Rio J) 2019; 95 Suppl 1:49-58. [PMID: 30593791 DOI: 10.1016/j.jped.2018.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 10/31/2018] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES This study aimed to review the literature on the repercussions of the different inborn errors of immunity on growth, drawing attention to the diagnosis of this group of diseases in patients with growth disorders, as well as to enable the identification of the different causes of growth disorders in patients with inborn errors of immunity, which can help in their treatment. DATA SOURCES Non-systematic review of the literature, searching articles since 2000 in PubMed with the terms "growth", "growth disorders", "failure to thrive", or "short stature" AND "immunologic deficiency syndromes", "immune deficiency disease", or "immune deficiency" NOT HIV. The Online Mendelian Inheritance in Man (OMIN) database was searched for immunodeficiencies and short stature or failure to thrive. DATA SUMMARY Inborn errors of immunity can affect growth in different ways, and some of them can change growth through multiple simultaneous mechanisms: genetic syndromes; disorders of the osteoarticular system; disorders of the endocrine system; reduction in caloric intake; catabolic processes; loss of nutrients; and inflammatory and/or infectious conditions. CONCLUSIONS The type of inborn errors of immunity allows anticipating what type of growth disorder can be expected. The type of growth disorder can help in the diagnosis of clinical conditions related to inborn errors of immunity. In many inborn errors of immunity, the causes of poor growth are mixed, involving more than one factor. In many cases, impaired growth can be adjusted with proper inborn errors of immunity treatment or proper approach to the mechanism of growth impairment.
Collapse
Affiliation(s)
- Ekaterini Simões Goudouris
- Universidade Federal do Rio de Janeiro (UFRJ), Faculdade de Medicina, Departamento de Pediatria, Rio de Janeiro, RJ, Brazil; Universidade Federal do Rio de Janeiro (UFRJ), Instituto de Puericultura e Pediatria Martagão Gesteira (IPPMG), Curso de Especialização em Alergia e Imunologia Clínica, Rio de Janeiro, RJ, Brazil.
| | - Gesmar Rodrigues Silva Segundo
- Universidade Federal de Uberlândia (UFU), Faculdade de Medicina, Departamento de Pediatria, Uberlândia, MG, Brazil; Universidade Federal de Uberlândia (UFU), Hospital das Clínicas, Programa de Residência Médica em Alergia e Imunologia Pediátrica, Uberlândia, MG, Brazil
| | - Cecilia Poli
- Universidad del Desarrollo, Facultad de Medicina, Instituto de Ciencias e Innovación em Medicina, Santiago, Chile
| |
Collapse
|
25
|
Clinical Aspects of STAT3 Gain-of-Function Germline Mutations: A Systematic Review. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2019; 7:1958-1969.e9. [PMID: 30825606 DOI: 10.1016/j.jaip.2019.02.018] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Signal transducer and activator of transcription 3 (STAT3) gain-of-function (GOF) germline mutations have been recently described. A comprehensive overview of this early-onset multiorgan autoimmune and lymphoproliferative disease has not yet been compiled. OBJECTIVE We have conducted a systematic review of published STAT3 GOF cases to describe clinical, diagnostic, and therapeutic aspects of the disease. METHODS A systematic review including articles published before October 10, 2018, in PubMed, Web of Science, and Cochrane Central Register of Controlled Trials databases was performed. We described cases of patients with STAT3 GOF germline mutations with genetic analysis and a concordant phenotype if functional analyses were not performed for the mutation. RESULTS The search identified 18 publications describing 42 unique patients. Twenty-eight different mutations were described. Onset of disease was very early with an average age of 3 (0.5-5) years. The most frequent manifestations were autoimmune cytopenias (28 of 42), lymphoproliferation (27 of 42), enteropathy (24 of 42), interstitial lung disease (15 of 42), thyroiditis (13 of 42), diabetes (10 of 42), and postnatal growth failure (15 of 21). Immunodeficiency was not always a predominant feature. Most patients required significant immunosuppressive therapy. Five patients received hematopoietic stem cell transplantation, and 4 died from complications. Improvement of symptoms was observed for 8 of 9 patients who received targeted biotherapies. CONCLUSIONS STAT3 GOF syndrome is a new clinical entity to consider when confronted with a patient with early-onset polyautoimmunity, lymphoproliferation, and growth failure. At this time, precise therapeutic guidelines are lacking, but use of anti-IL-6 receptor and JAK inhibitor biologics is an attractive possibility.
Collapse
|
26
|
Olbrich P, Freeman AF. STAT1 and STAT3 mutations: important lessons for clinical immunologists. Expert Rev Clin Immunol 2018; 14:1029-1041. [PMID: 30280610 DOI: 10.1080/1744666x.2018.1531704] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The transcription factors signal transducer and activator of transcription (STAT) 1 and STAT3 fulfill fundamental functions in nonimmune and immune cells. The description and follow-up of patients with germline mutations that result in either loss-of-function or gain-of-function have contributed to our understanding of the pathophysiology of these regulators. Depending on the type of mutations, clinical symptoms are complex and can include infection susceptibility, immune dysregulation as well as characteristic nonimmune features. Areas covered: In this review, we provide an overview about mechanistic concepts, clinical manifestations, diagnostic process, and traditional as well as innovative treatment options aiming to help the clinical immunologist to better understand and manage these complex and rare diseases. Clinical and research papers were identified and summarized through PubMed Internet searches, and expert opinions are provided. Expert commentary: The last several years have seen an explosion in the clinical descriptions and pathogenesis knowledge of the diseases caused by GOF and LOF mutations in STAT1 and STAT3. However, harmonization of laboratory testing and follow-up in international cohorts is needed to increase our knowledge about the natural history of these disorders as well as the development of curative or supportive targeted therapies.
Collapse
Affiliation(s)
- Peter Olbrich
- a Sección de Infectología, Reumatologíe e Inmunología Pediátrica (SIRIP) , Hospital Infantil Universitario Virgen del Rocío , Seville , Spain.,b Grupo de Enfermedades Infecciosas e Inmunodeficiencias , Instituto de Biomedicina de Sevilla (IBiS) , Seville , Spain
| | - Alexandra F Freeman
- c National Institute of Allergy and Infectious Diseases, NIH , Bethesda , MD , USA
| |
Collapse
|
27
|
Gutiérrez M, Scaglia P, Keselman A, Martucci L, Karabatas L, Domené S, Martin A, Pennisi P, Blanco M, Sanguineti N, Bezrodnik L, Di Giovanni D, Caldirola MS, Azcoiti ME, Gaillard MI, Denson LA, Zhang K, Husami A, Yayah Jones NH, Hwa V, Revale S, Vázquez M, Jasper H, Kumar A, Domené H. Partial growth hormone insensitivity and dysregulatory immune disease associated with de novo germline activating STAT3 mutations. Mol Cell Endocrinol 2018; 473:166-177. [PMID: 29378236 PMCID: PMC6143347 DOI: 10.1016/j.mce.2018.01.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/23/2018] [Accepted: 01/23/2018] [Indexed: 01/09/2023]
Abstract
Germinal heterozygous activating STAT3 mutations represent a novel monogenic defect associated with multi-organ autoimmune disease and, in some cases, severe growth retardation. By using whole-exome sequencing, we identified two novel STAT3 mutations, p.E616del and p.C426R, in two unrelated pediatric patients with IGF-I deficiency and immune dysregulation. The functional analyses showed that both variants were gain-of-function (GOF), although they were not constitutively phosphorylated. They presented differences in their dephosphorylation kinetics and transcriptional activities under interleukin-6 stimulation. Both variants increased their transcriptional activities in response to growth hormone (GH) treatment. Nonetheless, STAT5b transcriptional activity was diminished in the presence of STAT3 GOF variants, suggesting a disruptive role of STAT3 GOF variants in the GH signaling pathway. This study highlights the broad clinical spectrum of patients presenting activating STAT3 mutations and explores the underlying molecular pathway responsible for this condition, suggesting that different mutations may drive increased activity by slightly different mechanisms.
Collapse
Affiliation(s)
- Mariana Gutiérrez
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Paula Scaglia
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Ana Keselman
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Lucía Martucci
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Liliana Karabatas
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Sabina Domené
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Ayelen Martin
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Patricia Pennisi
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Miguel Blanco
- Endocrinología, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Nora Sanguineti
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Liliana Bezrodnik
- Inmunología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | | | | | | | - María Isabel Gaillard
- Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lee A Denson
- Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kejian Zhang
- Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ammar Husami
- Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nana-Hawa Yayah Jones
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vivian Hwa
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Santiago Revale
- Instituto de Agrobiotecnología de Rosario (INDEAR), CONICET, Rosario, Argentina
| | - Martín Vázquez
- Instituto de Agrobiotecnología de Rosario (INDEAR), CONICET, Rosario, Argentina
| | - Héctor Jasper
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Ashish Kumar
- Division of BM Transplantation and Immunodeficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Horacio Domené
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina.
| |
Collapse
|
28
|
Nunes-Santos CDJ, Rosenzweig SD. Bacille Calmette-Guerin Complications in Newly Described Primary Immunodeficiency Diseases: 2010-2017. Front Immunol 2018; 9:1423. [PMID: 29988375 PMCID: PMC6023996 DOI: 10.3389/fimmu.2018.01423] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/07/2018] [Indexed: 12/25/2022] Open
Abstract
Bacille Calmette–Guerin (BCG) vaccine is widely used as a prevention strategy against tuberculosis. BCG is a live vaccine, usually given early in life in most countries. While safe to most recipients, it poses a risk to immunocompromised patients. Several primary immunodeficiency diseases (PIDD) have been classically associated with complications related to BCG vaccine. However, a number of new inborn errors of immunity have been described lately in which little is known about adverse reactions following BCG vaccination. The aim of this review is to summarize the existing data on BCG-related complications in patients diagnosed with PIDD described since 2010. When BCG vaccination status or complications were not specifically addressed in those manuscripts, we directly contacted the corresponding authors for further clarification. We also analyzed data on other mycobacterial infections in these patients. Based on our analysis, around 8% of patients with gain-of-function mutations in STAT1 had mycobacterial infections, including localized complications in 3 and disseminated disease in 4 out of 19 BCG-vaccinated patients. Localized BCG reactions were also frequent in activated PI3Kδ syndrome type 1 (3/10) and type 2 (2/18) vaccinated children. Also, of note, no BCG-related complications have been described in either CTLA4 or LRBA protein-deficient patients; and not enough information on BCG-vaccinated NFKB1 or NFKB2-deficient patients was available to drive any conclusions about these diseases. Despite the high prevalence of environmental mycobacterial infections in GATA2-deficient patients, only one case of BCG reaction has been reported in a patient who developed disseminated disease. In conclusion, BCG complications could be expected in some particular, recently described PIDD and it remains a preventable risk factor for pediatric PIDD patients.
Collapse
Affiliation(s)
- Cristiane de Jesus Nunes-Santos
- Faculdade de Medicina, Instituto da Crianca, Universidade de São Paulo, São Paulo, Brazil.,Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
29
|
Cirillo F, Lazzeroni P, Catellani C, Sartori C, Amarri S, Street ME. MicroRNAs link chronic inflammation in childhood to growth impairment and insulin-resistance. Cytokine Growth Factor Rev 2018; 39:1-18. [DOI: 10.1016/j.cytogfr.2017.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 02/07/2023]
|