1
|
Szafron V, Shah SR, Holzmann-Pazgal G, Toledanes G, Rider NL. Recurrent Breast Abscesses in a Female with Autosomal Dominant Hyper-IgE Syndrome. J Clin Immunol 2022; 42:889-891. [PMID: 35292907 DOI: 10.1007/s10875-022-01248-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Vibha Szafron
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA. .,Section of Immunology, Allergy, and Retrovirology, Texas Children's Hospital, Houston, TX, USA.
| | - Sohail R Shah
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.,Division of Pediatric Surgery, Texas Children's Hospital, Houston, TX, USA
| | - Galit Holzmann-Pazgal
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.,Section of Infectious Diseases, Texas Children's Hospital, Houston, TX, USA
| | - Giancarlo Toledanes
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.,Section of Pediatric Hospital Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Nicholas L Rider
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.,Section of Immunology, Allergy, and Retrovirology, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
2
|
Development of vascular disease models to explore disease causation and pathomechanisms of rare vascular diseases. Semin Immunopathol 2022; 44:259-268. [PMID: 35233690 PMCID: PMC8887661 DOI: 10.1007/s00281-022-00925-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022]
Abstract
As the field of medicine is striving forward heralded by a new era of next-generation sequencing (NGS) and integrated technologies such as bioprinting and biological material development, the utility of rare monogenetic vascular disease modeling in this landscape is starting to emerge. With their genetic simplicity and broader applicability, these patient-specific models are at the forefront of modern personalized medicine. As a collective, rare diseases are a significant burden on global healthcare systems, and rare vascular diseases make up a significant proportion of this. High costs are due to a lengthy diagnostic process, affecting all ages from infants to adults, as well as the severity and chronic nature of the disease. Their complex nature requires sophisticated disease models and integrated approaches involving multidisciplinary teams. Here, we review these emerging vascular disease models, how they contribute to our understanding of the pathomechanisms in rare vascular diseases and provide useful platforms for therapeutic discovery.
Collapse
|
3
|
STAT3 is critical for skeletal development and bone homeostasis by regulating osteogenesis. Nat Commun 2021; 12:6891. [PMID: 34824272 PMCID: PMC8616950 DOI: 10.1038/s41467-021-27273-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 10/19/2021] [Indexed: 11/08/2022] Open
Abstract
Skeletal deformities are typical AD-HIES manifestations, which are mainly caused by heterozygous and loss-of-function mutations in Signal transducer and activator of transcription 3 (STAT3). However, the mechanism is still unclear and the treatment strategy is limited. Herein, we reported that the mice with Stat3 deletion in osteoblasts, but not in osteoclasts, induced AD-HIES-like skeletal defects, including craniofacial malformation, osteoporosis, and spontaneous bone fracture. Mechanistic analyses revealed that STAT3 in cooperation with Msh homeobox 1(MSX1) drove osteoblast differentiation by promoting Distal-less homeobox 5(Dlx5) transcription. Furthermore, pharmacological activation of STAT3 partially rescued skeletal deformities in heterozygous knockout mice, while inhibition of STAT3 aggravated bone loss. Taken together, these data show that STAT3 is critical for modulating skeletal development and maintaining bone homeostasis through STAT3-indcued osteogenesis and suggest it may be a potential target for treatments.
Collapse
|
4
|
Tsilifis C, Freeman AF, Gennery AR. STAT3 Hyper-IgE Syndrome-an Update and Unanswered Questions. J Clin Immunol 2021; 41:864-880. [PMID: 33932191 PMCID: PMC8249299 DOI: 10.1007/s10875-021-01051-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/20/2021] [Indexed: 12/16/2022]
Abstract
The hyper-IgE syndromes (HIES) are a heterogeneous group of inborn errors of immunity sharing manifestations including increased infection susceptibility, eczema, and raised serum IgE. Since the prototypical HIES description 55 years ago, areas of significant progress have included description of key disease-causing genes and differentiation into clinically distinct entities. The first two patients reported had what is now understood to be HIES from dominant-negative mutations in signal transduction and activator of transcription 3 (STAT3-HIES), conferring a broad immune defect across both innate and acquired arms, as well as defects in skeletal, connective tissue, and vascular function, causing a clinical phenotype including eczema, staphylococcal and fungal skin and pulmonary infection, scoliosis and minimal trauma fractures, and vascular tortuosity and aneurysm. Due to the constitutionally expressed nature of STAT3, initial reports at treatment with allogeneic stem cell transplantation were not positive and treatment has hinged on aggressive antimicrobial prophylaxis and treatment to prevent the development of end-organ disease such as pneumatocele. Research into the pathophysiology of STAT3-HIES has driven understanding of the interface of several signaling pathways, including the JAK-STAT pathways, interleukins 6 and 17, and the role of Th17 lymphocytes, and has been expanded by identification of phenocopies such as mutations in IL6ST and ZNF341. In this review we summarize the published literature on STAT3-HIES, present the diverse clinical manifestations of this syndrome with current management strategies, and update on the uncertain role of stem cell transplantation for this disease. We outline key unanswered questions for further study.
Collapse
Affiliation(s)
- Christo Tsilifis
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew R Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK.
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
5
|
Zhang S, Lei C, Wu J, Xiao M, Zhou J, Zhu S, Fu J, Lu D, Sun X, Xu C. A comprehensive and universal approach for embryo testing in patients with different genetic disorders. Clin Transl Med 2021; 11:e490. [PMID: 34323405 PMCID: PMC8265165 DOI: 10.1002/ctm2.490] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/01/2021] [Accepted: 06/20/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND In vitro fertilization (IVF) with preimplantation genetic testing (PGT) has markedly improved clinical pregnancy outcomes for carriers of gene mutations or chromosomal structural rearrangements by the selection of embryos free of disease-causing genes and chromosome abnormalities. However, for detecting whole or segmental chromosome aneuploidies, gene variants or balanced chromosome rearrangements in the same embryo require separate procedures, and none of the existing detection platforms is universal for all patients with different genetic disorders. METHODS Here, we report a cost-effective, family-based haplotype phasing approach that can simultaneously evaluate multiple genetic variants, including monogenic disorders, aneuploidy, and balanced chromosome rearrangements in the same embryo with a single test. A total of 12 monogenic diseases carrier couples and either of them carried chromosomal rearrangements were enrolled simultaneously in this present study. Genome-wide genotyping was performed with single-nucleotide polymorphism (SNP)-array, and aneuploidies were analyzed through SNP allele frequency and Log R ratio. Parental haplotypes were phased by an available genotype from a close relative, and the embryonic genome-wide haplotypes were determined through family haplotype linkage analysis (FHLA). Disease-causing genes and chromosomal rearrangements were detected by haplotypes located within the 2 Mb region covering the targeted genes or breakpoint regions. RESULTS Twelve blastocysts were thawed, and then transferred into the uterus of female patients. Nine pregnancies had reached the second trimester and five healthy babies have been born. Fetus validation results, performed with the amniotic fluid or umbilical cord blood samples, were consistent with those at the blastocyst stage diagnosed by PGT. CONCLUSIONS We demonstrate that SNP-based FHLA enables the accurate genetic detection of a wide spectrum of monogenic diseases and chromosome abnormalities in embryos, preventing the transfer of parental genetic abnormalities to the fetus. This method can be implemented as a universal platform for embryo testing in patients with different genetic disorders.
Collapse
Affiliation(s)
- Shuo Zhang
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| | - Caixia Lei
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| | - Junping Wu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| | - Min Xiao
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| | - Jing Zhou
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| | - Saijuan Zhu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| | - Jing Fu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| | - Daru Lu
- State Key Laboratory of Genetic Engineering, School of Life ScienceFudan UniversityShanghaiChina
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family PlanningScience and Technology Research InstituteChongqingChina
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
- Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| | - Congjian Xu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
- Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
| |
Collapse
|
6
|
Eberherr AC, Maaske A, Wolf C, Giesert F, Berutti R, Rusha E, Pertek A, Kastlmeier MT, Voss C, Plummer M, Sayed A, Graf E, Effner R, Volz T, Drukker M, Strom TM, Meitinger T, Stoeger T, Buyx AM, Hagl B, Renner ED. Rescue of STAT3 Function in Hyper-IgE Syndrome Using Adenine Base Editing. CRISPR J 2021; 4:178-190. [PMID: 33876960 DOI: 10.1089/crispr.2020.0111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
STAT3-hyper IgE syndrome (STAT3-HIES) is a primary immunodeficiency presenting with destructive lung disease along with other symptoms. CRISPR-Cas9-mediated adenine base editors (ABEs) have the potential to correct one of the most common STAT3-HIES causing heterozygous STAT3 mutations (c.1144C>T/p.R382W). As a proof-of-concept, we successfully applied ABEs to correct STAT3 p.R382W in patient fibroblasts and induced pluripotent stem cells (iPSCs). Treated primary STAT3-HIES patient fibroblasts showed a correction efficiency of 29% ± 7% without detectable off-target effects evaluated through whole-genome and high-throughput sequencing. Compared with untreated patient fibroblasts, corrected single-cell clones showed functional rescue of STAT3 signaling with significantly increased STAT3 DNA-binding activity and target gene expression of CCL2 and SOCS3. Patient-derived iPSCs were corrected with an efficiency of 30% ± 6% and differentiated to alveolar organoids showing preserved plasticity in treated cells. In conclusion, our results are supportive for ABE-based gene correction as a potential causative treatment of STAT3-HIES.
Collapse
Affiliation(s)
- Andreas C Eberherr
- Translational Immunology in Environmental Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Translational Immunology, Institute of Environmental Medicine, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Andre Maaske
- Translational Immunology in Environmental Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Translational Immunology, Institute of Environmental Medicine, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Christine Wolf
- Translational Immunology in Environmental Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Translational Immunology, Institute of Environmental Medicine, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Florian Giesert
- Institute of Developmental Genetics, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany.,Chair of Developmental Genetics, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany
| | - Riccardo Berutti
- Institute of Human Genetics, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany.,Institute of Neurogenomics, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Ejona Rusha
- iPSC Core Facility, Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Anna Pertek
- iPSC Core Facility, Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Miriam T Kastlmeier
- Institute of Lung Biology and Disease, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Carola Voss
- Institute of Lung Biology and Disease, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Michelle Plummer
- Translational Immunology in Environmental Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Translational Immunology, Institute of Environmental Medicine, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Amina Sayed
- Translational Immunology in Environmental Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Translational Immunology, Institute of Environmental Medicine, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany.,Institute of Lung Biology and Disease, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Elisabeth Graf
- Institute of Human Genetics, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Renate Effner
- Translational Immunology in Environmental Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Translational Immunology, Institute of Environmental Medicine, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Thomas Volz
- Department of Dermatology and Allergology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; and Technical University of Munich, Munich, Germany
| | - Micha Drukker
- iPSC Core Facility, Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Tim M Strom
- Institute of Human Genetics, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Tobias Stoeger
- Institute of Lung Biology and Disease, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Alena M Buyx
- Institute for History and Ethics of Medicine, Technical University of Munich, Munich, Germany
| | - Beate Hagl
- Translational Immunology in Environmental Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Translational Immunology, Institute of Environmental Medicine, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| | - Ellen D Renner
- Translational Immunology in Environmental Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Munich, Germany.,Translational Immunology, Institute of Environmental Medicine, Helmholtz Zentrum Munich, Neuherberg, Germany; Technical University of Munich, Munich, Germany
| |
Collapse
|
7
|
McCann KJ, Yadav M, Alishahedani ME, Freeman AF, Myles IA. Differential responses to folic acid in an established keloid fibroblast cell line are mediated by JAK1/2 and STAT3. PLoS One 2021; 16:e0248011. [PMID: 33662027 PMCID: PMC7932104 DOI: 10.1371/journal.pone.0248011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/17/2021] [Indexed: 11/22/2022] Open
Abstract
Keloids are a type of disordered scar formation which not only show heterogeneity between individuals and within the scar itself, but also share common features of hyperproliferation, abnormal extra-cellular matrix deposition and degradation, as well as altered expression of the molecular markers of wound healing. Numerous reports have established that cells from keloid scars display Warburg metabolism—a form of JAK2/STAT3-induced metabolic adaptation typical of rapidly dividing cells in which glycolysis becomes the predominant source of ATP over oxidative phosphorylation (OxPhos). Using the JAK1/2 inhibitor ruxolitinib, along with cells from patients with STAT3 loss of function (STA3 LOF; autosomal dominant hyper IgE syndrome) we examined the role of JAK/STAT signaling in the hyperproliferation and metabolic dysregulation seen in keloid fibroblasts. Although ruxolitinib inhibited hyperactivity in the scratch assay in keloid fibroblasts, it paradoxically exacerbated the hyper-glycolytic state, possibly by further limiting OxPhos via alterations in mitochondrial phosphorylated STAT3 (pSTAT3Ser727). In healthy volunteer fibroblasts, folic acid exposure recapitulated the exaggerated closure and hyper-glycolytic state of keloid fibroblasts through JAK1/2- and STAT3-dependent pathways. Although additional studies are needed before extrapolating from a representative cell line to keloids writ large, our results provide novel insights into the metabolic consequences of STAT3 dysfunction, suggest a possible role for folate metabolism in the pathogenesis of keloid scars, and offer in vitro pre-clinical data supporting considerations of clinical trials for ruxolitinib in keloid disorder.
Collapse
Affiliation(s)
- Katelyn J. McCann
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Manoj Yadav
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mohammadali E. Alishahedani
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Ian A. Myles
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
8
|
Mitchell AL, Urban AK, Freeman AF, Hammoud DA. An Unusual Pattern of Premature Cervical Spine Degeneration in STAT3-LOF. J Clin Immunol 2021; 41:576-584. [PMID: 33404973 DOI: 10.1007/s10875-020-00926-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/16/2020] [Indexed: 01/21/2023]
Abstract
Loss of function mutations in STAT3 (STAT3-LOF; autosomal dominant hyper-IgE (Job's) syndrome) are associated with a variety of musculoskeletal manifestations, including scoliosis, osteoporosis, and minimal trauma fractures. This retrospective magnetic resonance (MR) imaging study sought to characterize an unusual pattern of cervical spine degeneration among a cohort of STAT3-LOF patients. Cervical spine MR images of the STAT3-LOF cohort (n = 38) were assessed for a variety of degenerative changes and compared to age-matched groups of controls (n = 42) without known immune or musculoskeletal abnormalities. A unique pattern of premature cervical spine degeneration was identified among the STAT3-LOF cohort which included straightening and eventual reversal of the normal cervical lordosis, mainly due to multi-level spondylolisthesis, as well as early development of spinal canal narrowing, cord compression, and myelomalacia. Cervical spine degeneration in the STAT3-LOF cohort was significantly worse than controls in both the 30-45 and 45 + age groups. Moderate to severe degenerative changes were present after age 30, and markedly worsened over time in several cases. Bone mineral density (BMD) had a moderate negative correlation with cervical degeneration severity and a strong negative correlation with age among STAT3-LOF participants. Cervical degeneration in STAT3-LOF appears to be progressive and could result in cord compromise if left unaddressed. Focused history and physical examination for signs of neurologic compromise as well as periodic MR imaging are thus recommended for the evaluation of premature cervical spine degeneration in STAT3-LOF patients after age 30 so that timely surgical interventions may be considered to prevent spinal cord damage and permanent neurological deficits.
Collapse
Affiliation(s)
- Amelia L Mitchell
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Amanda K Urban
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Gough P, Myles IA. Tumor Necrosis Factor Receptors: Pleiotropic Signaling Complexes and Their Differential Effects. Front Immunol 2020; 11:585880. [PMID: 33324405 PMCID: PMC7723893 DOI: 10.3389/fimmu.2020.585880] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022] Open
Abstract
Since its discovery in 1975, TNFα has been a subject of intense study as it plays significant roles in both immunity and cancer. Such attention is well deserved as TNFα is unique in its engagement of pleiotropic signaling via its two receptors: TNFR1 and TNFR2. Extensive research has yielded mechanistic insights into how a single cytokine can provoke a disparate range of cellular responses, from proliferation and survival to apoptosis and necrosis. Understanding the intracellular signaling pathways induced by this single cytokine via its two receptors is key to further revelation of its exact functions in the many disease states and immune responses in which it plays a role. In this review, we describe the signaling complexes formed by TNFR1 and TNFR2 that lead to each potential cellular response, namely, canonical and non-canonical NF-κB activation, apoptosis and necrosis. This is followed by a discussion of data from in vivo mouse and human studies to examine the differential impacts of TNFR1 versus TNFR2 signaling.
Collapse
Affiliation(s)
- Portia Gough
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| | - Ian A Myles
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
10
|
Ponsford MJ, Clark J, Mock J, Abinun M, Carne E, El-Shanawany T, Williams PE, Choudhury A, Freeman AF, Gennery AR, Jolles S. Hematopoietic Stem Cell Transplantation and Vasculopathy Associated With STAT3-Dominant-Negative Hyper-IgE Syndrome. Front Pediatr 2020; 8:575. [PMID: 33014947 PMCID: PMC7511721 DOI: 10.3389/fped.2020.00575] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/06/2020] [Indexed: 02/04/2023] Open
Abstract
Dominant negative mutations in the transcription-factor STAT3 underlie the rare primary immunodeficiency Job's syndrome. Allogeneic Hematopoietic Stem Cell Transplantation (HSCT) has shown promise in correction of the underlying immunological defect, with one report suggesting HSCT can prevent development of wider connective tissue complications. Here, we report the case of a 26 year old male who developed an acute ST-elevation myocardial infarction due to coronary artery ectasia and thrombosis, occurring despite pediatric allogeneic HSCT for STAT3-HIES and a predicted 10-year conventional cardiovascular risk of 0.1%. Vasculopathy associated with STAT3-HIES may persist or arise following HSCT and can precipitate life-threatening complications. This has implications for counseling and vascular surveillance, and highlights the need for further studies to determine the risk, pathogenesis, and optimal management of the vasculopathy associated with STAT3-HIES.
Collapse
Affiliation(s)
- Mark J. Ponsford
- Immunodeficiency Centre for Wales, University Hospital for Wales, Cardiff, United Kingdom
- Division of Infection, Inflammation, and Immunity, School of Medicine, Tenovus Institute, Cardiff University, Cardiff, United Kingdom
| | - James Clark
- Department of Interventional Cardiology, University Hospital for Wales, Cardiff, United Kingdom
| | - Joel Mock
- Department of Interventional Cardiology, University Hospital for Wales, Cardiff, United Kingdom
| | - Mario Abinun
- Paediatric Haematopoietic Stem Cell Transplant Unit, Translational and Clinical Research Institute, Great North Children's Hospital, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Emily Carne
- Immunodeficiency Centre for Wales, University Hospital for Wales, Cardiff, United Kingdom
| | - Tariq El-Shanawany
- Immunodeficiency Centre for Wales, University Hospital for Wales, Cardiff, United Kingdom
| | - Paul E. Williams
- Immunodeficiency Centre for Wales, University Hospital for Wales, Cardiff, United Kingdom
| | - Anirban Choudhury
- Department of Interventional Cardiology, Morriston Hospital, Swansea Bay University Local Health Board, Swansea, United Kingdom
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Andrew R. Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Translational and Clinical Research Institute, Great North Children's Hospital, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital for Wales, Cardiff, United Kingdom
| |
Collapse
|