1
|
Barbetti F, Deeb A, Suzuki S. Neonatal diabetes mellitus around the world: Update 2024. J Diabetes Investig 2024; 15:1711-1724. [PMID: 39344692 PMCID: PMC11615689 DOI: 10.1111/jdi.14312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024] Open
Abstract
Neonatal diabetes mellitus (NDM), defined as diabetes with an onset during the first 6 months of life, is a rare form of monogenic diabetes. The initial publications on this condition began appearing in the second half of the 1990s and quite surprisingly, the search for new NDM genes is still ongoing with great vigor. Between 2018 and early 2024, six brand new NDM-genes have been discovered (CNOT1, FICD, ONECUT1, PDIA6, YIPF5, ZNF808) and three genes known to cause different diseases were identified as NDM-genes (EIF2B1, NARS2, KCNMA1). In addition, NDM cases carrying mutations in three other genes known to give rise to diabetes during childhood have been also identified (AGPAT2, BSCL2, PIK3R1). As a consequence, the list of NDM genes now exceeds 40. This genetic heterogeneity translates into many different mechanism(s) of disease that are being investigated with state-of-the-art methodologies, such as induced pluripotent stem cells (iPSC) and human embryonic stem cells (hESC) manipulated with the CRISPR technique of genome editing. This diversity in genetic causes and the pathophysiology of diabetes dictate the need for a variety of therapeutic approaches. The aim of this paper is to provide an overview on recent achievements in all aspects of this area of research.
Collapse
Affiliation(s)
- Fabrizio Barbetti
- Monogenic Diabetes Clinic, Endocrinology and Diabetes UnitBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Asma Deeb
- Pediatric Endocrine Division, Sheikh Shakhbout Medical City and College of Medicine and Health ScienceKhalifa UniversityAbu DhabiUAE
| | - Shigeru Suzuki
- Department of PediatricsAsahikawa Medical UniversityAsahikawaJapan
| |
Collapse
|
2
|
Rapini N, Delvecchio M, Mucciolo M, Ruta R, Rabbone I, Cherubini V, Zucchini S, Cianfarani S, Prandi E, Schiaffini R, Bizzarri C, Piccini B, Maltoni G, Predieri B, Minuto N, Di Paola R, Giordano M, Tinto N, Grasso V, Russo L, Tiberi V, Scaramuzza A, Frontino G, Maggio MC, Musolino G, Piccinno E, Tinti D, Carrera P, Mozzillo E, Cappa M, Iafusco D, Bonfanti R, Novelli A, Barbetti F. The Changing Landscape of Neonatal Diabetes Mellitus in Italy Between 2003 and 2022. J Clin Endocrinol Metab 2024; 109:2349-2357. [PMID: 38408297 PMCID: PMC11319002 DOI: 10.1210/clinem/dgae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/29/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024]
Abstract
CONTEXT In the last decade the Sanger method of DNA sequencing has been replaced by next-generation sequencing (NGS). NGS is valuable in conditions characterized by high genetic heterogeneity such as neonatal diabetes mellitus (NDM). OBJECTIVE To compare results of genetic analysis of patients with NDM and congenital severe insulin resistance (c.SIR) identified in Italy in 2003-2012 (Sanger) vs 2013-2022 (NGS). METHODS We reviewed clinical and genetic records of 104 cases with diabetes onset before 6 months of age (NDM + c.SIR) of the Italian dataset. RESULTS Fifty-five patients (50 NDM + 5 c.SIR) were identified during 2003-2012 and 49 (46 NDM + 3 c.SIR) in 2013-2022. Twenty-year incidence was 1:103 340 (NDM) and 1:1 240 082 (c.SIR) live births. Frequent NDM/c.SIR genetic defects (KCNJ11, INS, ABCC8, 6q24, INSR) were detected in 41 and 34 probands during 2003-2012 and 2013-2022, respectively. We identified a pathogenic variant in rare genes in a single proband (GATA4) (1/42 or 2.4%) during 2003-2012 and in 8 infants (RFX6, PDX1, GATA6, HNF1B, FOXP3, IL2RA, LRBA, BSCL2) during 2013-2022 (8/42 or 19%, P = .034 vs 2003-2012). Notably, among rare genes 5 were recessive. Swift and accurate genetic diagnosis led to appropriate treatment: patients with autoimmune NDM (FOXP3, IL2RA, LRBA) were subjected to bone marrow transplant; patients with pancreas agenesis/hypoplasia (RFX6, PDX1) were supplemented with pancreatic enzymes, and the individual with lipodystrophy caused by BSCL2 was started on metreleptin. CONCLUSION NGS substantially improved diagnosis and precision therapy of monogenic forms of neonatal diabetes and c.SIR in Italy.
Collapse
Affiliation(s)
- Novella Rapini
- Monogenic Diabetes Clinic, Endocrinology and Diabetes Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Maurizio Delvecchio
- Metabolic Disorder and Diabetes Unit, “Giovanni XXIII” Children Hospital, 70100 Bari, Italy
- Unit of Pediatrics, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mafalda Mucciolo
- Translational Cytogenomics Research Unit, Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Rosario Ruta
- Translational Cytogenomics Research Unit, Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Ivana Rabbone
- Department of Health Sciences, Division of Pediatrics, University of Eastern Piedmont, 28100 Novara, Italy
| | - Valentino Cherubini
- Pediatric Endocrinology and Diabetology Unit, Department of Women's and Children's Health, Azienda Ospedaliero Universitaria delle Marche, G. Salesi Hospital, 60126 Ancona, Italy
| | - Stefano Zucchini
- Pediatric Endocrine Unit, University Hospital of Bologna Sant’Orsola-Malpighi, 40138 Bologna, Italy
| | - Stefano Cianfarani
- Endocrinology and Diabetes Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Elena Prandi
- Pediatrics Clinic, University of Brescia and ASST Spedali Civili of Brescia, 25123 Brescia, Italy
| | - Riccardo Schiaffini
- Endocrinology and Diabetes Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Carla Bizzarri
- Endocrinology and Diabetes Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Barbara Piccini
- Endocrinology and Diabetology Unit, Meyer University Children's Hospital IRCCS, 50139 Florence, Italy
| | - Giulio Maltoni
- Pediatric Endocrine Unit, University Hospital of Bologna Sant’Orsola-Malpighi, 40138 Bologna, Italy
| | - Barbara Predieri
- Department of Medical and Surgical Sciences of Mother, Children and Adults, Pediatric Unit, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Nicola Minuto
- Regional Center for Pediatric Diabetes, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Rossella Di Paola
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Mara Giordano
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
- Laboratory of Genetics, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Nadia Tinto
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II/CEINGE Advanced Biotechnologies Franco Salvatore, 80131 Naples, Italy
| | - Valeria Grasso
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Lucia Russo
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valentina Tiberi
- Pediatric Endocrinology and Diabetology Unit, Department of Women's and Children's Health, Azienda Ospedaliero Universitaria delle Marche, G. Salesi Hospital, 60126 Ancona, Italy
| | - Andrea Scaramuzza
- Diabetes and Endocrine Service, Pediatric Unit, ASST Cremona, Maggiore Hospital, 26100 Cremona, Italy
| | - Giulio Frontino
- Department of Pediatrics, Pediatric Diabetology Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | | | - Gianluca Musolino
- Growth Disorders, Endocrinology and Diabetology Clinic, Filippo del Ponte Pediatric Hospital, ASST Sette Laghi, 21100 Varese, Italy
| | - Elvira Piccinno
- Metabolic Disorder and Diabetes Unit, “Giovanni XXIII” Children Hospital, 70100 Bari, Italy
| | - Davide Tinti
- Department of Pediatrics, University of Turin, 10126 Turin, Italy
| | - Paola Carrera
- Genomics for the Diagnosis of Human Pathologies, San Raffaele Scientific Institute, Center for Omics sciences @OSR, 20132 Milan, Italy
- Laboratory of Molecular Genetics and Cytogenetics, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Enza Mozzillo
- Department of Translational Medical Science, Section of Pediatrics, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Marco Cappa
- Research Area for Innovative Therapies in Endocrinopathies, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Dario Iafusco
- Department of Pediatrics, University of Campania Luigi Vanvitelli, 81100 Naples, Italy
| | - Riccardo Bonfanti
- Department of Pediatrics, Pediatric Diabetology Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele and Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Fabrizio Barbetti
- Monogenic Diabetes Clinic, Endocrinology and Diabetes Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
3
|
Pérez-Pérez D, Fuentes-Pananá EM, Flores-Hermenegildo JM, Romero-Ramirez H, Santos-Argumedo L, Kilimann MW, Rodríguez-Alba JC, Lopez-Herrera G. Lipopolysaccharide-responsive beige-like anchor is involved in regulating NF-κB activation in B cells. Front Immunol 2024; 15:1409434. [PMID: 39076990 PMCID: PMC11284061 DOI: 10.3389/fimmu.2024.1409434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/21/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Lipopolysaccharide-responsive and beige-like anchor (LRBA) is a scaffolding protein that interacts with proteins such as CTLA-4 and PKA, the importance of which has been determined in various cell types, including T regulatory cells, B cells, and renal cells. LRBA deficiency is associated with an inborn error in immunity characterized by immunodeficiency and autoimmunity. In addition to defects in T regulatory cells, patients with LRBA deficiency also exhibit B cell defects, such as reduced cell number, low memory B cells, hypogammaglobulinemia, impaired B cell proliferation, and increased autophagy. Although Lrba-/- mice do not exhibit the immunodeficiency observed in humans, responses to B cell receptors (BCR) in B cells have not been explored. Therefore, a murine model is for elucidating the mechanism of Lrba mechanism in B cells. Aim To compare and evaluate spleen-derived B cell responses to BCR crosslinking in C57BL6 Lrba-/- and Lrba+/+ mice. Materials and methods Spleen-derived B cells were obtained from 8 to 12-week-old mice. Subpopulations were determined by immunostaining and flow cytometry. BCR crosslinking was assessed by the F(ab')2 anti-μ chain. Activation, proliferation and viability assays were performed using flow cytometry and protein phosphorylation was evaluated by immunoblotting. The nuclear localization of p65 was determined using confocal microscopy. Nur77 expression was evaluated by Western blot. Results Lrba-/- B cells showed an activated phenotype and a decreased proportion of transitional 1 B cells, and both proliferation and survival were affected after BCR crosslinking in the Lrba-/- mice. The NF-κB pathway exhibited a basal activation status of several components, resulting in increased activation of p50, p65, and IκBα, basal p50 activation was reduced by the Plcγ2 inhibitor U73122. BCR crosslinking in Lrba-/ - B cells resulted in poor p50 phosphorylation and p65 nuclear localization. Increased levels of Nur77 were detected. Discussion These results indicate the importance of Lrba in controlling NF-κB activation driven by BCR. Basal activation of NF-κB could impact cellular processes, such as, activation, differentiation, proliferation, and maintenance of B cells after antigen encounter.
Collapse
Affiliation(s)
- Daniela Pérez-Pérez
- Doctorate Program in Biological Sciences, Autonomous National University of Mexico, Mexico City, Mexico
- Immunodeficiency Laboratory, National Institute of Pediatrics, Mexico City, Mexico
| | | | - José Mizael Flores-Hermenegildo
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV IPN, Mexico City, Mexico
| | - Hector Romero-Ramirez
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV IPN, Mexico City, Mexico
| | - Leopoldo Santos-Argumedo
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV IPN, Mexico City, Mexico
| | - Manfred W. Kilimann
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Juan Carlos Rodríguez-Alba
- Medicine and Surgery Faculty, Autonomous University Benito Juarez from Oaxaca, Oaxaca, Mexico
- Neuroimmunology and Neurooncology Unit, The National Institute of Neurology and Neurosurgery (NINN), Mexico City, Mexico
| | | |
Collapse
|
4
|
Perez-Perez D, Santos-Argumedo L, Rodriguez-Alba JC, Lopez-Herrera G. Analysis of LRBA pathogenic variants and the association with functional protein domains and clinical presentation. Pediatr Allergy Immunol 2024; 35:e14179. [PMID: 38923448 DOI: 10.1111/pai.14179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/29/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
LRBA is a cytoplasmic protein that is ubiquitously distributed. Almost all LRBA domains have a scaffolding function. In 2012, it was reported that homozygous variants in LRBA are associated with early-onset hypogammaglobulinemia. Since its discovery, more than 100 pathogenic variants have been reported. This review focuses on the variants reported in LRBA and their possible associations with clinical phenotypes. In this work LRBA deficiency cases reported more than 11 years ago have been revised. A database was constructed to analyze the type of variants, age at onset, clinical diagnosis, infections, autoimmune diseases, and cellular and immunoglobulin levels. The review of cases from 2012 to 2023 showed that LRBA deficiency was commonly diagnosed in patients with a clinical diagnosis of Common Variable Immunodeficiency, followed by enteropathy, neonatal diabetes mellitus, ALPS, and X-linked-like syndrome. Most cases show early onset of presentation at <6 years of age. Most cases lack protein expression, whereas hypogammaglobulinemia is observed in half of the cases, and IgG and IgA levels are isotypes reported at low levels. Patients with elevated IgG levels exhibited more than one autoimmune manifestation. Patients carrying pathogenic variants leading to a premature stop codon show a severe phenotype as they have an earlier onset of disease presentation, severe autoimmune manifestations, premature death, and low B cells and regulatory T cell levels. Missense variants were more common in patients with low IgG levels and cytopenia. This work lead to the conclusion that the type of variant in LRBA has association with disease severity, which leads to a premature stop codon being the ones that correlates with severe disease.
Collapse
Affiliation(s)
- D Perez-Perez
- Doctorate Program in Biological Sciences, Autonomous National University of Mexico, Mexico City, Mexico
- Immunodeficiencies Laboratory, National Institute of Pediatrics (INP), Mexico City, Mexico
| | - L Santos-Argumedo
- Biomedicine Department, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | - J C Rodriguez-Alba
- Neuroimmunology and Neurooncology Unit, The National Institute of Neurology and Neurosurgery (NINN), Mexico City, Mexico
- Medicine and Surgery Faculty, Autonomous University Benito Juarez from Oaxaca, Oaxaca, Mexico
| | - G Lopez-Herrera
- Immunodeficiencies Laboratory, National Institute of Pediatrics (INP), Mexico City, Mexico
| |
Collapse
|
5
|
López-Nevado M, Ortiz-Martín J, Serrano C, Pérez-Saez MA, López-Lorenzo JL, Gil-Etayo FJ, Rodríguez-Frías E, Cabrera-Marante O, Morales-Pérez P, Rodríguez-Pinilla MS, Manso R, Salgado-Sánchez RN, Cerdá-Montagud A, Quesada-Espinosa JF, Gómez-Rodríguez MJ, Paz-Artal E, Muñoz-Calleja C, Arranz-Sáez R, Allende LM. Novel Germline TET2 Mutations in Two Unrelated Patients with Autoimmune Lymphoproliferative Syndrome-Like Phenotype and Hematologic Malignancy. J Clin Immunol 2023; 43:165-180. [PMID: 36066697 DOI: 10.1007/s10875-022-01361-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/26/2022] [Indexed: 01/18/2023]
Abstract
Somatic mutations in the ten-eleven translocation methylcytosine dioxygenase 2 gene (TET2) have been associated to hematologic malignancies. More recently, biallelic, and monoallelic germline mutations conferring susceptibility to lymphoid and myeloid cancer have been described. We report two unrelated autoimmune lymphoproliferative syndrome-like patients who presented with T-cell lymphoma associated with novel germline biallelic or monoallelic mutations in the TET2 gene. Both patients presented a history of chronic lymphoproliferation with lymphadenopathies and splenomegaly, cytopenias, and immune dysregulation. We identified the first compound heterozygous patient for TET2 mutations (P1) and the first ALPS-like patient with a monoallelic TET2 mutation (P2). P1 had the most severe form of autosomal recessive disease due to TET2 loss of function resulting in absent TET2 expression and profound increase in DNA methylation. Additionally, the immunophenotype showed some alterations in innate and adaptive immune system as inverted myeloid/plasmacytoid dendritic cells ratio, elevated terminally differentiated effector memory CD8 + T-cells re-expressing CD45RA, regulatory T-cells, and Th2 circulating follicular T-cells. Double-negative T-cells, vitamin B12, and IL-10 were elevated according to the ALPS-like suspicion. Interestingly, the healthy P1's brother carried a TET2 mutation and presented some markers of immune dysregulation. P2 showed elevated vitamin B12, hypergammaglobulinemia, and decreased HDL levels. Therefore, novel molecular defects in TET2 confirm and expand both clinical and immunological phenotype, contributing to a better knowledge of the bridge between cancer and immunity.
Collapse
Affiliation(s)
- Marta López-Nevado
- Immunology Department, University Hospital 12 de Octubre, Av de Córdoba s/n, 28041, Madrid, Spain.
- Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain.
| | | | - Cristina Serrano
- Immunology Department, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - María A Pérez-Saez
- Hematology Department, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - José L López-Lorenzo
- Hematology Department, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Francisco J Gil-Etayo
- Immunology Department, University Hospital 12 de Octubre, Av de Córdoba s/n, 28041, Madrid, Spain
- Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Edgar Rodríguez-Frías
- Immunology Department, University Hospital 12 de Octubre, Av de Córdoba s/n, 28041, Madrid, Spain
- Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Oscar Cabrera-Marante
- Immunology Department, University Hospital 12 de Octubre, Av de Córdoba s/n, 28041, Madrid, Spain
- Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Pablo Morales-Pérez
- Immunology Department, University Hospital 12 de Octubre, Av de Córdoba s/n, 28041, Madrid, Spain
- Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Rebeca Manso
- Pathology Department, Research Institute Fundación Jiménez Díaz, Madrid, Spain
| | | | - Ana Cerdá-Montagud
- Hematology Department, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Juan F Quesada-Espinosa
- Genetics Department, University Hospital 12 de Octubre, Madrid, Spain
- UDisGen (Unidad de Dismorfología Y Genética), University Hospital 12 de Octubre, Madrid, Spain
| | - María J Gómez-Rodríguez
- Genetics Department, University Hospital 12 de Octubre, Madrid, Spain
- UDisGen (Unidad de Dismorfología Y Genética), University Hospital 12 de Octubre, Madrid, Spain
| | - Estela Paz-Artal
- Immunology Department, University Hospital 12 de Octubre, Av de Córdoba s/n, 28041, Madrid, Spain
- Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- School of Medicine, Complutense University of Madrid, Madrid, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Immunology Department, University Hospital La Princesa, Madrid, Spain
- School of Medicine, University Autónoma de Madrid, Madrid, Spain
- Research Institute Hospital de La Princesa, Madrid, Spain
| | - Reyes Arranz-Sáez
- Hematology Department, University Hospital La Princesa, Madrid, Spain
| | - Luis M Allende
- Immunology Department, University Hospital 12 de Octubre, Av de Córdoba s/n, 28041, Madrid, Spain.
- Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain.
- School of Medicine, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
6
|
Harley ITW, Allison K, Scofield RH. Polygenic autoimmune disease risk alleles impacting B cell tolerance act in concert across shared molecular networks in mouse and in humans. Front Immunol 2022; 13:953439. [PMID: 36090990 PMCID: PMC9450536 DOI: 10.3389/fimmu.2022.953439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Most B cells produced in the bone marrow have some level of autoreactivity. Despite efforts of central tolerance to eliminate these cells, many escape to periphery, where in healthy individuals, they are rendered functionally non-responsive to restimulation through their antigen receptor via a process termed anergy. Broad repertoire autoreactivity may reflect the chances of generating autoreactivity by stochastic use of germline immunoglobulin gene segments or active mechanisms may select autoreactive cells during egress to the naïve peripheral B cell pool. Likewise, it is unclear why in some individuals autoreactive B cell clones become activated and drive pathophysiologic changes in autoimmune diseases. Both of these remain central questions in the study of the immune system(s). In most individuals, autoimmune diseases arise from complex interplay of genetic risk factors and environmental influences. Advances in genome sequencing and increased statistical power from large autoimmune disease cohorts has led to identification of more than 200 autoimmune disease risk loci. It has been observed that autoantibodies are detectable in the serum years to decades prior to the diagnosis of autoimmune disease. Thus, current models hold that genetic defects in the pathways that control autoreactive B cell tolerance set genetic liability thresholds across multiple autoimmune diseases. Despite the fact these seminal concepts were developed in animal (especially murine) models of autoimmune disease, some perceive a disconnect between human risk alleles and those identified in murine models of autoimmune disease. Here, we synthesize the current state of the art in our understanding of human risk alleles in two prototypical autoimmune diseases - systemic lupus erythematosus (SLE) and type 1 diabetes (T1D) along with spontaneous murine disease models. We compare these risk networks to those reported in murine models of these diseases, focusing on pathways relevant to anergy and central tolerance. We highlight some differences between murine and human environmental and genetic factors that may impact autoimmune disease development and expression and may, in turn, explain some of this discrepancy. Finally, we show that there is substantial overlap between the molecular networks that define these disease states across species. Our synthesis and analysis of the current state of the field are consistent with the idea that the same molecular networks are perturbed in murine and human autoimmune disease. Based on these analyses, we anticipate that murine autoimmune disease models will continue to yield novel insights into how best to diagnose, prognose, prevent and treat human autoimmune diseases.
Collapse
Affiliation(s)
- Isaac T. W. Harley
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
- Rheumatology Section, Medicine Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Kristen Allison
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
| | - R. Hal Scofield
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Medical/Research Service, US Department of Veterans Affairs Medical Center, Oklahoma City, OK, United States
| |
Collapse
|
7
|
Hawari I, Haris B, Mohammed I, Ericsson J, Khalifa A, Hussain K. Infancy onset diabetes mellitus in a patient with a novel homozygous LRBA mutation. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY CASE REPORTS 2022. [DOI: 10.1016/j.jecr.2022.100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|