1
|
McDonald GB, Landsverk OJ, McGovern DP, Aasebø A, Paulsen V, Haritunians T, Reims HM, McLaughlin BM, Zisman T, Li D, Elholm ET, Jahnsen FL, Georges GE, Gedde-Dahl T. Allogeneic bone marrow transplantation for patients with treatment-refractory Crohn's Disease. Heliyon 2024; 10:e24026. [PMID: 38283244 PMCID: PMC10818189 DOI: 10.1016/j.heliyon.2024.e24026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/16/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Background & aims Durable remissions of Crohn's Disease (CD) have followed myeloablative conditioning therapy and allogeneic marrow transplantation. For patients with treatment-refractory disease, we used reduced-intensity conditioning to minimize toxicity, marrow from donors with low Polygenic Risk Scores for CD as cell sources, and protracted immune suppression to lower the risk of graft-versus-host disease (GVHD). Our aim was to achieve durable CD remissions while minimizing transplant-related complications. Methods DNA from patients and their HLA-matched unrelated donors was genotyped and Polygenic Risk Scores calculated. Donor marrow was infused following non-myeloablative conditioning. Patient symptoms and endoscopic findings were documented at intervals after transplant. Results We screened 807 patients, 143 of whom met eligibility criteria; 2 patients received allografts. Patient 1 had multiple complications and died at day 332 from respiratory failure. Patient 2 had resolution of CD symptoms until day 178 when CD recurred, associated with persistent host chimerism in both peripheral blood and intestinal mucosa. Withdrawal of immune suppression was followed by dominant donor immune chimerism in peripheral blood and resolution of CD findings. Over time, mucosal T-cells became donor-dominant. At 5 years after allografting, Patient 2 remained off all medications but had mild symptoms related to a jejunal stricture that required stricturoplasty at 6 years. At 8 years, she remains stable off medications. Conclusions The kinetics of immunologic chimerism after allogeneic marrow transplantation for CD patients depends on the intensity of the conditioning regimen and the magnitude of immune suppression. One patient achieved durable improvement of her previously refractory CD only after establishing donor immunologic chimerism in intestinal mucosa. Her course provides proof-of-principal for allografting as a potential treatment for refractory CD, but an immunoablative conditioning regimen should be considered for future studies.(ClinicalTrials.gov, NCT01570348).
Collapse
Affiliation(s)
- George B. McDonald
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Dermot P.B. McGovern
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anders Aasebø
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Vemund Paulsen
- Department of Transplantation Medicine, Section of Gastroenterology, Oslo University Hospital Rikshospitalet, Norway
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Henrik M. Reims
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | | | - Timothy Zisman
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Dalin Li
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Elisabeth T.M.M. Elholm
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Hematology, Oslo University Hospital, Norway and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Frode L. Jahnsen
- Department of Pathology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Norway
| | - George E. Georges
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Tobias Gedde-Dahl
- Department of Hematology, Oslo University Hospital, Norway and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Lee J, Sim KM, Kang M, Oh HJ, Choi HJ, Kim YE, Pack CG, Kim K, Kim KM, Oh SH, Kim I, Chang I. Understanding the molecular mechanism of pathogenic variants of BIR2 domain in XIAP-deficient inflammatory bowel disease. Sci Rep 2024; 14:853. [PMID: 38191507 PMCID: PMC10774423 DOI: 10.1038/s41598-023-50932-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/28/2023] [Indexed: 01/10/2024] Open
Abstract
X-linked inhibitor of apoptosis protein (XIAP) deficiency causes refractory inflammatory bowel disease. The XIAP protein plays a pivotal role in the pro-inflammatory response through the nucleotide-binding oligomerization domain-containing signaling pathway that is important in mucosal homeostasis. We analyzed the molecular mechanism of non-synonymous pathogenic variants (PVs) of XIAP BIR2 domain. We generated N-terminally green fluorescent protein-tagged XIAP constructs of representative non-synonymous PVs. Co-immunoprecipitation and fluorescence cross-correlation spectroscopy showed that wild-type XIAP and RIP2 preferentially interacted in live cells, whereas all non-synonymous PV XIAPs failed to interact properly with RIP2. Structural analysis showed that various structural changes by mutations, such as hydrophobic core collapse, Zn-finger loss, and spatial rearrangement, destabilized the two loop structures (174-182 and 205-215) that critically interact with RIP2. Subsequently, it caused a failure of RIP2 ubiquitination and loss of protein deficiency by the auto-ubiquitination of all XIAP mutants. These findings could enhance our understanding of the role of XIAP mutations in XIAP-deficient inflammatory bowel disease and may benefit future therapeutic strategies.
Collapse
Affiliation(s)
- Juhwan Lee
- iProtein Therapeutics Inc., Munji-ro 281-9, Yuseong-gu, Daejeon, Korea
| | - Kyoung Mi Sim
- Department of Convergence Medicine, Asan Medical Center, Asan Institutes for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Mooseok Kang
- iProtein Therapeutics Inc., Munji-ro 281-9, Yuseong-gu, Daejeon, Korea
| | - Hyun Ju Oh
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Ho Jung Choi
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Yeong Eun Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine, Asan Medical Center, Asan Institutes for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyunggon Kim
- Department of Convergence Medicine, Asan Medical Center, Asan Institutes for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Mo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Seak Hee Oh
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-Gu, Seoul, 05505, Korea.
| | - Inki Kim
- Department of Convergence Medicine, Asan Medical Center, Asan Institutes for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea.
- Department of Pharmacology, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-Gu, Seoul, 05505, Korea.
| | - Iksoo Chang
- Creative Research Initiatives Center for Proteome Biophysics, Department of Brain Sciences and Supercomputing Bigdata Center, DGIST, Daegu, 42988, Korea.
- Department of Brain Sciences and Supercomputing Big Data Center, DGIST, Daegu, 42988, Korea.
| |
Collapse
|
3
|
Lethier M, Huard K, Hons M, Favier A, Brutscher B, Boeri Erba E, Abbott DW, Cusack S, Pellegrini E. Structure shows that the BIR2 domain of E3 ligase XIAP binds across the RIPK2 kinase dimer interface. Life Sci Alliance 2023; 6:e202201784. [PMID: 37673444 PMCID: PMC10485824 DOI: 10.26508/lsa.202201784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023] Open
Abstract
RIPK2 is an essential adaptor for NOD signalling and its kinase domain is a drug target for NOD-related diseases, such as inflammatory bowel disease. However, recent work indicates that the phosphorylation activity of RIPK2 is dispensable for signalling and that inhibitors of both RIPK2 activity and RIPK2 ubiquitination prevent the essential interaction between RIPK2 and the BIR2 domain of XIAP, the key RIPK2 ubiquitin E3 ligase. Moreover, XIAP BIR2 antagonists also block this interaction. To reveal the molecular mechanisms involved, we combined native mass spectrometry, NMR, and cryo-electron microscopy to determine the structure of the RIPK2 kinase BIR2 domain complex and validated the interface with in cellulo assays. The structure shows that BIR2 binds across the RIPK2 kinase antiparallel dimer and provides an explanation for both inhibitory mechanisms. It also highlights why phosphorylation of the kinase activation loop is dispensable for signalling while revealing the structural role of RIPK2-K209 residue in the RIPK2-XIAP BIR2 interaction. Our results clarify the features of the RIPK2 conformation essential for its role as a scaffold protein for ubiquitination.
Collapse
Affiliation(s)
| | - Karine Huard
- European Molecular Biology Laboratory, Grenoble, France
| | - Michael Hons
- European Molecular Biology Laboratory, Grenoble, France
| | - Adrien Favier
- University Grenoble Alpes, IBS, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Bernhard Brutscher
- University Grenoble Alpes, IBS, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Elisabetta Boeri Erba
- University Grenoble Alpes, IBS, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Derek W Abbott
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | | | | |
Collapse
|
4
|
Hughes SA, Lin M, Weir A, Huang B, Xiong L, Chua NK, Pang J, Santavanond JP, Tixeira R, Doerflinger M, Deng Y, Yu C, Silke N, Conos SA, Frank D, Simpson DS, Murphy JM, Lawlor KE, Pearson JS, Silke J, Pellegrini M, Herold MJ, Poon IKH, Masters SL, Li M, Tang Q, Zhang Y, Rashidi M, Geng L, Vince JE. Caspase-8-driven apoptotic and pyroptotic crosstalk causes cell death and IL-1β release in X-linked inhibitor of apoptosis (XIAP) deficiency. EMBO J 2023; 42:e110468. [PMID: 36647737 PMCID: PMC9975961 DOI: 10.15252/embj.2021110468] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
Genetic lesions in X-linked inhibitor of apoptosis (XIAP) pre-dispose humans to cell death-associated inflammatory diseases, although the underlying mechanisms remain unclear. Here, we report that two patients with XIAP deficiency-associated inflammatory bowel disease display increased inflammatory IL-1β maturation as well as cell death-associated caspase-8 and Gasdermin D (GSDMD) processing in diseased tissue, which is reduced upon patient treatment. Loss of XIAP leads to caspase-8-driven cell death and bioactive IL-1β release that is only abrogated by combined deletion of the apoptotic and pyroptotic cell death machinery. Namely, extrinsic apoptotic caspase-8 promotes pyroptotic GSDMD processing that kills macrophages lacking both inflammasome and apoptosis signalling components (caspase-1, -3, -7, -11 and BID), while caspase-8 can still cause cell death in the absence of both GSDMD and GSDME when caspase-3 and caspase-7 are present. Neither caspase-3 and caspase-7-mediated activation of the pannexin-1 channel, or GSDMD loss, prevented NLRP3 inflammasome assembly and consequent caspase-1 and IL-1β maturation downstream of XIAP inhibition and caspase-8 activation, even though the pannexin-1 channel was required for NLRP3 triggering upon mitochondrial apoptosis. These findings uncouple the mechanisms of cell death and NLRP3 activation resulting from extrinsic and intrinsic apoptosis signalling, reveal how XIAP loss can co-opt dual cell death programs, and uncover strategies for targeting the cell death and inflammatory pathways that result from XIAP deficiency.
Collapse
|
5
|
Ruan J, Schlüter D, Naumann M, Waisman A, Wang X. Ubiquitin-modifying enzymes as regulators of colitis. Trends Mol Med 2022; 28:304-318. [PMID: 35177326 DOI: 10.1016/j.molmed.2022.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 12/18/2022]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is a chronic inflammatory disorder of the gastrointestinal tract. Although the pathophysiology of IBD is multifaceted, ubiquitination, a post-translational modification, has been shown to have essential roles in its pathogenesis and development. Ubiquitin-modifying enzymes (UMEs) work in synergy to orchestrate the optimal ubiquitination of target proteins, thereby maintaining intestinal homeostasis. Genome-wide association studies (GWAS) have identified multiple UME genes as IBD susceptibility loci, implying the importance of UMEs in IBD. Furthermore, accumulative evidence demonstrates that UMEs affect intestinal inflammation by regulating various aspects, such as intestinal barrier functions and immune responses. Considering the significant functions of UMEs in IBD, targeting UMEs could become a favorable therapeutic approach for IBD.
Collapse
Affiliation(s)
- Jing Ruan
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Xu Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China; Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
6
|
Yang L, Booth C, Speckmann C, Seidel MG, Worth AJ, Kindle G, Lankester AC, B G, Gennery AR, Seppanen MR, Morris EC, Burns SO. Phenotype, genotype, treatment, and survival outcomes in patients with X-linked inhibitor of apoptosis deficiency. J Allergy Clin Immunol 2021; 150:456-466. [PMID: 34920033 DOI: 10.1016/j.jaci.2021.10.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/06/2021] [Accepted: 10/13/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND X-linked inhibitor of apoptosis (XIAP) deficiency is a rare, primary immunodeficiency disease caused by XIAP gene mutations. A broad range of phenotype, severity, and age of onset present challenges for patient management. OBJECTIVE To characterize the phenotype, treatment, and survival outcomes of XIAP deficiency and assess parameters influencing prognosis. METHODS Data published from 2006-2020 were retrospectively analyzed. RESULTS 167 patients from 117 families with XIAP deficiency were reported with 90 different mutations. A wide spectrum of clinical features were seen, of which hemophagocytic lymphohistiocytosis (HLH) and inflammatory bowel disease (IBD) were the most common. Patients frequently developed multiple features with no clear genotype-phenotype correlation. 117 patients were managed conservatively and 50 underwent hematopoietic stem cell transplantation (HSCT), with respective overall survival probabilities of 90% and 53% at age 16 years. The predominant indication for HSCT was early-onset HLH. Active HLH and myeloablative conditioning regimens increased HSCT-related mortality, although HSCT outcome was much better after 2015 than before. For conservatively managed patients reaching adulthood, survival probabilities were 86% at age 30 years and 37% by age 52 years, with worse outcomes for patients developing the disease before the age of 5 years or with new disease features in adulthood. 9 asymptomatic mutation carriers were identified with a median age of 13.5 years. CONCLUSIONS Our study demonstrates the variable nature of XIAP deficiency which evolves over life for individual patients. Better therapeutic strategies and prospective studies are required to reduce morbidity and mortality and improve decision-making and long-term outcomes for patients with XIAP deficiency.
Collapse
Affiliation(s)
- Linlin Yang
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London NW3 2PF, United Kingdom; Institute for Immunity and Transplantation, University College London, London NW3 2PF, United Kingdom; Department of Hematology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Claire Booth
- Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Trust, London WC1N 1JH; Molecular and Cellular Immunology, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Carsten Speckmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, Medical Center - University of Freiburg, Germany; Center for Pediatrics and Adolescent Medicine, Department of Pediatric Hematology and Oncology, Faculty of Medicine, Medical Center - University of Freiburg, Germany
| | - Markus G Seidel
- Research Unit for Pediatric Hematology and Immunology, Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Austen Jj Worth
- Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Trust, London WC1N 1JH
| | - Gerhard Kindle
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, Medical Center - University of Freiburg, Germany
| | - Arjan C Lankester
- Willem-Alexander Children's Hospital, Department of Pediatrics, Stem Cell Transplantation program, Leiden University Medical Center, Leiden, The Netherlands
| | - Grimbacher B
- Institute for Immunity and Transplantation, University College London, London NW3 2PF, United Kingdom; Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, Medical Center - University of Freiburg, Germany; DZIF - German Center for Infection Research, Satellite Center Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, Albert-Ludwigs University, Freiburg, Germany; RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Germany
| | | | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University and Pediatric Immunology + HSCT, Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Mikko Rj Seppanen
- HUS Rare Disease Center, Children and Adolescents, University of Helsinki and Helsinki University Hospital, Finland
| | - Emma C Morris
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London NW3 2PF, United Kingdom; Institute for Immunity and Transplantation, University College London, London NW3 2PF, United Kingdom
| | - Siobhan O Burns
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London NW3 2PF, United Kingdom; Institute for Immunity and Transplantation, University College London, London NW3 2PF, United Kingdom.
| |
Collapse
|
7
|
Bachmann J, Le Thi G, Brückner A, Kalteis AL, Schwerd T, Koletzko S, Lurz E. Epstein-Barr Virus Prevalence at Diagnosis and Seroconversion during Follow-Up in Pediatric Inflammatory Bowel Disease. J Clin Med 2021; 10:jcm10215187. [PMID: 34768707 PMCID: PMC8584287 DOI: 10.3390/jcm10215187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
Primary Epstein-Barr virus infection in pediatric patients with inflammatory bowel disease during immunomodulation with thiopurines has been associated with increased risk for malignancies or hemophagocytic lymphohistiocytosis. We determined Epstein-Barr virus (EBV) seroprevalence at inflammatory bowel disease (IBD) diagnosis and seroconversion during follow-up in a large single center cohort of children with IBD. EBV serology results and patient characteristics were retrospectively retrieved from the hospital documentation system. EBV seronegative patients at IBD diagnosis were prospectively retested. We report on IBD patients with symptomatic active EBV infection and a complicated disease course, and those diagnosed with malignancy with respect to EBV status and drug exposure. Of 402 patients, 194 (48%) had available EBV serology results at time of IBD diagnosis at a median of 12 years (IQR 9-14 years). Thereof, 102 (53%) were EBV-positive. Of 92 EBV-negative patients, 66 were retested and 17% showed a seroconversion at a mean follow-up time of 4.3 years (SD 3 years). Three children treated with azathioprine experienced acute clinically relevant EBV infection 2, 2.5, and 4 years after IBD diagnosis, two developed signs of hemophagocytic lymphohistiocytosis. Three cases of malignancy occurred in the cohort, though none seemed to be triggered by EBV. In conclusion, almost 50% of pediatric IBD patients were EBV-naïve following diagnosis and may be at increased risk to develop severe EBV infection during immunosuppressive therapy, potentially associated with complications such as hemophagocytic lymphohistiocytosis or malignancy.
Collapse
Affiliation(s)
- Jennifer Bachmann
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany; (J.B.); (G.L.T.); (A.B.); (T.S.); (S.K.)
| | - Giang Le Thi
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany; (J.B.); (G.L.T.); (A.B.); (T.S.); (S.K.)
| | - Annecarin Brückner
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany; (J.B.); (G.L.T.); (A.B.); (T.S.); (S.K.)
| | - Anna-Lena Kalteis
- Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, 80336 Munich, Germany;
- German Center for Infection Research (DZIF), Partner Site Munich, 38124 Braunschweig, Germany
| | - Tobias Schwerd
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany; (J.B.); (G.L.T.); (A.B.); (T.S.); (S.K.)
| | - Sibylle Koletzko
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany; (J.B.); (G.L.T.); (A.B.); (T.S.); (S.K.)
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Collegium Medicum University of Warmia and Mazury, 10-561 Olsztyn, Poland
| | - Eberhard Lurz
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany; (J.B.); (G.L.T.); (A.B.); (T.S.); (S.K.)
- Correspondence: ; Tel.: +49-89-4400-519060
| |
Collapse
|
8
|
Chang I, Park S, Lee HJ, Kim I, Park S, Ahn MK, Lee J, Kang M, Baek IJ, Sung YH, Pack CG, Kang HJ, Lee K, Im HJ, Seo EJ, Kim KM, Yang SK, Song K, Oh SH. Interpretation of XIAP Variants of Uncertain Significance in Paediatric Patients with Refractory Crohn's Disease. J Crohns Colitis 2021; 15:1291-1304. [PMID: 33460440 DOI: 10.1093/ecco-jcc/jjab013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Mutations in XIAP can lead to the development of treatment-refractory severe paediatric Crohn's disease [CD], for which haematopoietic stem cell transplantation is the primary therapeutic option. The interpretation of variants of uncertain significance [VUSs] in XIAP needs to be scrutinized. METHODS Targeted next-generation sequencing was performed for 33 male paediatric patients with refractory CD admitted at a tertiary referral hospital. To obtain functional data, biomolecular cell assays and supercomputing molecular dynamics simulations were performed. RESULTS Nine unrelated male patients harboured hemizygous XIAP variants. Four known pathogenic variants and one novel pathogenic variant [p.Lys168Serfs*12] were identified in five patients, and two novel VUSs [p.Gly205del and p.Pro260Ser] and one known VUS [p.Glu350del] were identified in the remaining four. Among children with VUSs, only the subject with p.Gly205del exhibited defective NOD2 signalling. Using molecular dynamics simulation, we determined that the altered backbone torsional energy of C203 in XIAP of p.G205del was ~2 kcal/mol, suggesting loss of zinc binding in the mutant XIAP protein and poor coordination between the mutant XIAP and RIP2 proteins. Elevated auto-ubiquitination of zinc-depleted p.G205del XIAP protein resulted in XIAP protein deficiency. CONCLUSION A high prevalence of XIAP deficiency was noted among children with refractory CD. Advanced functional studies decreased the subjectivity in the case-level interpretation of XIAP VUSs and directed consideration of haematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Iksoo Chang
- Supercomputing & Big Data Center, DGIST, Daegu, Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, Korea
| | - Seongjun Park
- Department of Emerging Materials Science, DGIST, Daegu, Korea
| | - Hye-Jin Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Inki Kim
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sojung Park
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Kyoung Ahn
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Juhwan Lee
- Supercomputing & Big Data Center, DGIST, Daegu, Korea
| | - Mooseok Kang
- Department of Brain and Cognitive Sciences, DGIST, Daegu, Korea
| | - In-Jeoung Baek
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Hoon Sung
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyo-Jeong Kang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kunsong Lee
- Department of Pediatrics, Dankook University College of Medicine, Dankook University Hospital, Chungnam, Korea
| | - Ho Joon Im
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Eul Ju Seo
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Mo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyuyoung Song
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Korea
| | - Seak Hee Oh
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Speir M, Djajawi TM, Conos SA, Tye H, Lawlor KE. Targeting RIP Kinases in Chronic Inflammatory Disease. Biomolecules 2021; 11:biom11050646. [PMID: 33924766 PMCID: PMC8146010 DOI: 10.3390/biom11050646] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic inflammatory disorders are characterised by aberrant and exaggerated inflammatory immune cell responses. Modes of extrinsic cell death, apoptosis and necroptosis, have now been shown to be potent drivers of deleterious inflammation, and mutations in core repressors of these pathways underlie many autoinflammatory disorders. The receptor-interacting protein (RIP) kinases, RIPK1 and RIPK3, are integral players in extrinsic cell death signalling by regulating the production of pro-inflammatory cytokines, such as tumour necrosis factor (TNF), and coordinating the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome, which underpin pathological inflammation in numerous chronic inflammatory disorders. In this review, we firstly give an overview of the inflammatory cell death pathways regulated by RIPK1 and RIPK3. We then discuss how dysregulated signalling along these pathways can contribute to chronic inflammatory disorders of the joints, skin, and gastrointestinal tract, and discuss the emerging evidence for targeting these RIP kinases in the clinic.
Collapse
Affiliation(s)
- Mary Speir
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Tirta M. Djajawi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Stephanie A. Conos
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Hazel Tye
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
| | - Kate E. Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
- Correspondence: ; Tel.: +61-85722700
| |
Collapse
|
10
|
Gut Microbiota-Host Interactions in Inborn Errors of Immunity. Int J Mol Sci 2021; 22:ijms22031416. [PMID: 33572538 PMCID: PMC7866830 DOI: 10.3390/ijms22031416] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Inborn errors of immunity (IEI) are a group of disorders that are mostly caused by genetic mutations affecting immune host defense and immune regulation. Although IEI present with a wide spectrum of clinical features, in about one third of them various degrees of gastrointestinal (GI) involvement have been described and for some IEI the GI manifestations represent the main and peculiar clinical feature. The microbiome plays critical roles in the education and function of the host's innate and adaptive immune system, and imbalances in microbiota-immunity interactions can contribute to intestinal pathogenesis. Microbial dysbiosis combined to the impairment of immunosurveillance and immune dysfunction in IEI, may favor mucosal permeability and lead to inflammation. Here we review how immune homeostasis between commensals and the host is established in the gut, and how these mechanisms can be disrupted in the context of primary immunodeficiencies. Additionally, we highlight key aspects of the first studies on gut microbiome in patients affected by IEI and discuss how gut microbiome could be harnessed as a therapeutic approach in these diseases.
Collapse
|
11
|
Parackova Z, Milota T, Vrabcova P, Smetanova J, Svaton M, Freiberger T, Kanderova V, Sediva A. Novel XIAP mutation causing enhanced spontaneous apoptosis and disturbed NOD2 signalling in a patient with atypical adult-onset Crohn's disease. Cell Death Dis 2020; 11:430. [PMID: 32514016 PMCID: PMC7280281 DOI: 10.1038/s41419-020-2652-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022]
Abstract
X-linked inhibitor of apoptosis (XIAP) is the most potent human inhibitor of apoptosis, and is also involved in NOD2-dependent NFκB and MAPK signalling cascade activation. The absence or defective function of XIAP leads to the development of a rare and severe primary immunodeficiency known as X-linked lymphoproliferative syndrome type 2 (XLP-2), which is characterized by a triad of clinical manifestations, including a high incidence of haemophagocytic lymphohistiocytosis (HLH), lymphoproliferation and inflammatory bowel disease (IBD), usually with very early onset. Here, we present a novel XIAP mutation identified in a patient with atypical adult-onset IBD complicated by relapsing HLH, splenomegaly and sarcoid-like disease. The c.266delA mutation in the XIAP gene creates a premature stop codon, and causes a severe reduction in XIAP protein expression. The mutation is also associated with impaired spontaneous and staurosporine- and PMA-induced apoptosis accompanied by significantly increased expression of pro-apoptotic genes. We also confirmed the negative impact of this particular XIAP mutation on NOD2-dependent NFκB and MAPK activation, while NOD2-independent activation was found to be unaffected. Moreover, we assume that the mutation has an impact on the overproduction of IL-12 and IFNγ, the shift towards the Th1 immune response and increased numbers of central memory and effector memory CD4+ and CD8+ T cells. All these changes contribute to immune dysregulation and the clinical manifestation of XLP-2.
Collapse
Affiliation(s)
- Zuzana Parackova
- Department of Immunology, 2nd Faculty of Medicine Charles University, University Hospital in Motol, V Uvalu 84, Prague, Czech Republic.
| | - Tomas Milota
- Department of Immunology, 2nd Faculty of Medicine Charles University, University Hospital in Motol, V Uvalu 84, Prague, Czech Republic
| | - Petra Vrabcova
- Department of Immunology, 2nd Faculty of Medicine Charles University, University Hospital in Motol, V Uvalu 84, Prague, Czech Republic
| | - Jitka Smetanova
- Department of Immunology, 2nd Faculty of Medicine Charles University, University Hospital in Motol, V Uvalu 84, Prague, Czech Republic
| | - Michael Svaton
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Tomas Freiberger
- Molecular Genetics Laboratory, Center of Cardiovascular Surgery and Transplantation, Brno, Czech Republic.,Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Veronika Kanderova
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Anna Sediva
- Department of Immunology, 2nd Faculty of Medicine Charles University, University Hospital in Motol, V Uvalu 84, Prague, Czech Republic
| |
Collapse
|
12
|
Lega S, Pin A, Arrigo S, Cifaldi C, Girardelli M, Bianco AM, Malamisura M, Angelino G, Faraci S, Rea F, Romeo EF, Aloi M, Romano C, Barabino A, Martelossi S, Tommasini A, Di Matteo G, Cancrini C, De Angelis P, Finocchi A, Bramuzzo M. Diagnostic Approach to Monogenic Inflammatory Bowel Disease in Clinical Practice: A Ten-Year Multicentric Experience. Inflamm Bowel Dis 2020; 26:720-727. [PMID: 31375816 DOI: 10.1093/ibd/izz178] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS Multiple monogenic disorders present as very early onset inflammatory bowel disease (VEO-IBD) or as IBD with severe and atypical features. Establishing a genetic diagnosis may change patients' management and prognosis. In this study, we describe the diagnostic approach to suspected monogenic IBD in a real clinical setting, discussing genetic and phenotypic findings and therapeutic implications of molecular diagnosis. METHODS Information of patients with VEO-IBD and early onset IBD with severe/atypical phenotypes (EO-IBD s/a) managed between 2008-2017 who underwent a genetic workup were collected. RESULTS Ninety-three patients were included, and 12 (13%) reached a genetic diagnosis. Candidate sequencing (CS) was performed in 47 patients (50%), and next generation sequencing (NGS) was performed in 84 patients (90%). Candidate sequencing had a good diagnostic performance only when guided by clinical features specific for known monogenic diseases, whereas NGS helped finding new causative genetic variants and would have anticipated one monogenic diagnosis (XIAP) and consequent bone marrow transplant (BMT). Patients with monogenic IBD more frequently were male (92% vs 54%; P = 0.02), had extraintestinal findings (100% vs 34%; P < 0.001), and had disease onset ≤1 month of life (25% vs 1%; P = 0.006). Genetic diagnosis impacted patient management in 11 patients (92%), 7 of whom underwent BMT. CONCLUSION A genetic diagnosis can be established in a significant proportion of suspected monogenic IBD and has an impact on patients' management. Candidate sequencing may be deployed when clinical findings orientate toward a specific diagnosis. Next generation sequencing should be preferred in patients with nonspecific phenotypes.
Collapse
Affiliation(s)
- Sara Lega
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Alessia Pin
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Serena Arrigo
- Pediatric Gastroenterology and Endoscopy Unit, Institute Giannina Gaslini, Genoa, Italy
| | - Cristina Cifaldi
- Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Martina Girardelli
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Anna Monica Bianco
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Monica Malamisura
- Digestive Diseases Unit, Children's Hospital Bambino Gesù, Rome, Italy
| | - Giulia Angelino
- Digestive Diseases Unit, Children's Hospital Bambino Gesù, Rome, Italy
| | - Simona Faraci
- Digestive Diseases Unit, Children's Hospital Bambino Gesù, Rome, Italy
| | - Francesca Rea
- Digestive Diseases Unit, Children's Hospital Bambino Gesù, Rome, Italy
| | | | - Marina Aloi
- Pediatric Gastroenterology And Liver Unit, Sapienza University of Rome, Rome, Italy
| | - Claudio Romano
- Pediatric Gastroenterology and Cystic Fibrosis Unit, University of Messina, Messina, Italy
| | - Arrigo Barabino
- Pediatric Gastroenterology and Endoscopy Unit, Institute Giannina Gaslini, Genoa, Italy
| | - Stefano Martelossi
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Alberto Tommasini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | | | - Caterina Cancrini
- Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Paola De Angelis
- Digestive Diseases Unit, Children's Hospital Bambino Gesù, Rome, Italy
| | - Andrea Finocchi
- Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Matteo Bramuzzo
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
13
|
Chen T, Zhu C, Ye C. LncRNA CYTOR attenuates sepsis-induced myocardial injury via regulating miR-24/XIAP. Cell Biochem Funct 2020; 38:976-985. [PMID: 32181504 DOI: 10.1002/cbf.3524] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/04/2020] [Accepted: 01/20/2020] [Indexed: 02/06/2023]
Abstract
This work aims to investigate the function and mechanism of long non-coding RNA (lncRNA) cytoskeleton regulator RNA (CYTOR) in myocardial injury induced by sepsis. The sepsis-induced myocardial injury model in mice was established by intraperitoneal injection of LPS (10 mg/kg) in vivo, and cardiomyocyte H9c2 was treated with LPS to mimic sepsis in vitro. CYTOR expression and miR-24 expression were detected by qRT-PCR. After up-regulation or down-regulation of CYTOR and miR-24 expression in the H9c2 cells, and the viability of the cells was detected via MTT assay, and cell apoptosis was detected by TUNEL assay. Western blot was applied to determine the expression level of caspase 3, Bax and X-chromosome-linked inhibitor of apoptosis (XIAP). Interaction between CYTOR and miR-24 was determined by bioinformatics analysis, RT-PCR and dual luciferase reporter assay. Interaction between miR-24 and XIAP was determined through bioinformatics analysis, RT-PCR, western blot and dual luciferase reporter assay. CYTOR was markedly down-regulated. CYTOR interacted with miR-24, and negatively regulated its expression level. Over-expression of CYTOR or transfection of miR-24 inhibitors significantly promoted viability and inhibited apoptosis of H9c2 cells, while the knockdown of CYTOR and transfection of miR-24 mimics had opposite effects. CYTOR suppressed the expression level of apoptosis-related proteins, but miR-24 increased them. miR-24 directly targeted the 3'UTR of XIAP, and suppressed it, and XIAP was modulated indirectly by CYTOR. Down-regulation of CYTOR aggravates sepsis-induced cardiac injury via regulating miR-24 and XIAP.
Collapse
Affiliation(s)
- Ting Chen
- Intensive Care Unit, The Second People's Hospital of Heifei, Hefei, China
| | - Chunyan Zhu
- Intensive Care Unit, Anhui Provincial Hospital, the First Affiliated Hospital of University of Science and Technology of China, Heifei, China
| | - Chongyang Ye
- Intensive Care Unit, Anhui Provincial Hospital, the First Affiliated Hospital of University of Science and Technology of China, Heifei, China
| |
Collapse
|
14
|
Abstract
There are now 354 inborn errors of immunity (primary immunodeficiency diseases (PIDDs)) with 344 distinct molecular etiologies reported according to the International Union of Immunological Sciences (IUIS) (Clin Gastroenterol Hepatol 11: p. 1050-63, 2013, Semin Gastrointest Dis 8: p. 22-32, 1997, J Clin Immunol 38: p. 96-128, 2018). Using the IUIS document as a reference and cross-checking PubMed ( www.ncbi.nlm.nih.pubmed.gov ), we found that approximately one third of the 354 diseases of impaired immunity have a gastrointestinal component [J Clin Immunol 38: p. 96-128, 2018]. Often, the gastrointestinal symptomatology and pathology is the heralding sign of a PIDD; therefore, it is important to recognize patterns of disease which may manifest along the gastrointestinal tract as a more global derangement of immune function. As such, holistic consideration of immunity is warranted in patients with clinically significant gastrointestinal disease. Here, we discuss the manifold presentations and GI-specific complications of PIDDs which could lead patients to seek advice from a variety of clinician specialists. Often, patients with these medical problems will engage general pediatricians, surgeons, gastroenterologists, rheumatologists, and clinical immunologists among others. Following delineation of the presenting concern, accurate and often molecular diagnosis is imperative and a multi-disciplinary approach warranted for optimal management. In this review, we will summarize the current state of understanding of PIDD gastrointestinal disease involvement. We will do so by focusing upon gastrointestinal disease categories (i.e., inflammatory, diarrhea, nodular lymphoid hyperplasia, liver/biliary tract, structural disease, and oncologic disease) with an intent to aid the healthcare provider who may encounter a patient with an as-yet undiagnosed PIDD who presents initially with a gastrointestinal symptom, sign, or problem.
Collapse
|
15
|
Jensen S, Seidelin JB, LaCasse EC, Nielsen OH. SMAC mimetics and RIPK inhibitors as therapeutics for chronic inflammatory diseases. Sci Signal 2020; 13:13/619/eaax8295. [PMID: 32071170 DOI: 10.1126/scisignal.aax8295] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
New therapeutic approaches for chronic inflammatory diseases such as inflammatory bowel disease, rheumatoid arthritis, and psoriasis are needed because current treatments are often suboptimal in terms of both efficacy and the risks of serious adverse events. Inhibitor of apoptosis proteins (IAPs) are E3 ubiquitin ligases that inhibit cell death pathways and are themselves inhibited by second mitochondria-derived activator of caspases (SMAC). SMAC mimetics (SMs), small-molecule antagonists of IAPs, are being evaluated as cancer therapies in clinical trials. IAPs are also crucial regulators of inflammatory pathways because they influence both the activation of inflammatory genes and the induction of cell death through the receptor-interacting serine-threonine protein kinases (RIPKs), nuclear factor κB (NF-κB)-inducing kinase, and mitogen-activated protein kinases (MAPKs). Furthermore, there is an increasing interest in specifically targeting the substrates of IAP-mediated ubiquitylation, especially RIPK1, RIPK2, and RIPK3, as druggable nodes in inflammation control. Several studies have revealed an anti-inflammatory potential of RIPK inhibitors that either block inflammatory signaling or block the form of inflammatory cell death known as necroptosis. Expanding research on innate immune signaling through pattern recognition receptors that stimulate proinflammatory NF-κB and MAPK signaling may further contribute to uncovering the complex molecular roles used by IAPs and downstream RIPKs in inflammatory signaling. This may benefit and guide the development of SMs or selective RIPK inhibitors as anti-inflammatory therapeutics for various chronic inflammatory conditions.
Collapse
Affiliation(s)
- Simone Jensen
- Department of Gastroenterology, Medical Section, Herlev Hospital, University of Copenhagen, 1 Borgmester Ib Juuls Vej, DK-2730 Herlev, Denmark
| | - Jakob Benedict Seidelin
- Department of Gastroenterology, Medical Section, Herlev Hospital, University of Copenhagen, 1 Borgmester Ib Juuls Vej, DK-2730 Herlev, Denmark.
| | - Eric Charles LaCasse
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Medical Section, Herlev Hospital, University of Copenhagen, 1 Borgmester Ib Juuls Vej, DK-2730 Herlev, Denmark
| |
Collapse
|
16
|
Lekbua A, Ouahed J, O’Connell AE, Kahn SA, Goldsmith JD, Imamura T, Duncan CN, Kelsen JR, Worthey E, Snapper SB, Softic S. Risk-factors Associated With Poor Outcomes in VEO-IBD Secondary to XIAP Deficiency: A Case Report and Literature Review. J Pediatr Gastroenterol Nutr 2019; 69:e13-e18. [PMID: 31232887 PMCID: PMC6607918 DOI: 10.1097/mpg.0000000000002297] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Very early onset inflammatory bowel disease (VEO-IBD) represents a diagnostic and treatment challenge. Here we present a case of VEO-IBD secondary to a mutation in BIRC4 gene, which encodes X-linked inhibitor of apoptosis protein (XIAP), in a 17-month-old boy with severe failure to thrive, intractable diarrhea, and hepatosplenomegaly. Endoscopy and histology identified only mild duodenitis and ileitis, but severe pancolitis with crypt abscesses and epithelium apoptosis. Minimal improvement in symptoms was achieved with total parenteral nutrition (TPN), intravenous (IV) corticosteroids, and tacrolimus, whereas induction and maintenance therapy with adalimumab led to complete remission. After 6 months, the patient developed hemophagocytic lymphohistiocytosis and eventually died due to multisystem organ failure. A review of the literature revealed that some patients with VEO-IBD secondary to XIAP deficiency develop symptoms that are refractory to medical and surgical management, while initial reports suggest that allogeneic hematopoietic stem cell transplantation (HSCT), with reduced intensity conditioning, can successfully induce long-lasting remission and may even be curative. We propose that in patients with XIAP deficiency a constellation of symptoms including colitis at an early age, severe failure to thrive, and splenomegaly/hepatosplenomegaly can identify a subgroup of patients at high risk of experiencing medically refractory IBD phenotype and increased mortality. Hematopoietic stem cell transplant should be considered early in these high-risk patients, as it may resolve both their intestinal inflammation and a risk of developing life threatening hemophagocytic lymphohistiocytosis .
Collapse
Affiliation(s)
- Asama Lekbua
- Northeastern University, College of Science, Department of
Biology, Boston, MA
| | - Jodie Ouahed
- Division of Gastroenterology, Hepatology and Nutrition,
Boston Children’s Hospital, Boston, MA
| | - Amy E. O’Connell
- Division of Newborn Medicine, Boston Children’s
Hospital, Boston, MA
| | - Stacy A. Kahn
- Division of Gastroenterology, Hepatology and Nutrition,
Boston Children’s Hospital, Boston, MA
| | | | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of
Medicine, Kyoto, Japan
| | | | - Judith R. Kelsen
- Division of Gastroenterology, Hepatology and Nutrition,
Children’s Hospital of Philadelphia, Perelman School of Medicine, University
of Pennsylvania, Philadelphia, PA
| | | | - Scott B. Snapper
- Division of Gastroenterology, Hepatology and Nutrition,
Boston Children’s Hospital, Boston, MA
- Division of Gastroenterology, Hepatology & Endoscopy,
Brigham & Women’s Hospital, Boston MA
| | - Samir Softic
- Division of Gastroenterology, Hepatology and Nutrition,
Boston Children’s Hospital, Boston, MA
- Section on Integrative Physiology and Metabolism, Joslin
Diabetes Center, Boston, MA
| |
Collapse
|
17
|
Hrdinka M, Yabal M. Inhibitor of apoptosis proteins in human health and
disease. Genes Immun 2019; 20:641-650. [DOI: 10.1038/s41435-019-0078-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/23/2019] [Accepted: 04/01/2019] [Indexed: 12/13/2022]
|
18
|
Shabani M, Razaghian A, Alimadadi H, Shiari R, Shahrooei M, Parvaneh N. Different phenotypes of the same XIAP mutation in a family: A case of XIAP deficiency with juvenile idiopathic arthritis. Pediatr Blood Cancer 2019; 66:e27593. [PMID: 30604482 DOI: 10.1002/pbc.27593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/13/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Mahsima Shabani
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Anahita Razaghian
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran
| | - Hosein Alimadadi
- Department of Pediatric Gastroeneterology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shiari
- Department of Pediatric Rheumatology, Mofid Children Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Shahrooei
- Department of Microbiology and Immunology, Laboratory of Clinical Bacteriology and Mycology, KU Leuven, Leuven, Belgium.,Specialized Immunology Laboratory, Ahvaz, Iran
| | - Nima Parvaneh
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Oh SH, Sung YH, Kim I, Sim CK, Lee JH, Baek M, Pack CG, Seok C, Seo EJ, Lee MS, Kim KM. Novel Compound Heterozygote Mutation in IL10RA in a Patient With Very Early-Onset Inflammatory Bowel Disease. Inflamm Bowel Dis 2019; 25:498-509. [PMID: 30462267 DOI: 10.1093/ibd/izy353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Very early-onset inflammatory bowel disease (VEO-IBD) is often associated with monogenetic disorders. IL-10RA deficiency is one of the major causal mutations in VEO-IBD. Here, we aimed to identify the causal mutation associated with severe IBD in a 1-year-old patient, validate the pathogenicity of the mutation, and characterize the mutant protein. METHODS To identify the causal mutation, targeted exome sequencing (ES) was performed using the genomic DNA from the patient. To validate the pathogenicity, IL-10RA functional tests were performed using the patient's peripheral blood mononuclear cells (PBMCs). Additionally, flow cytometry analysis, confocal microscopy on overexpressed green fluorescent protein-fused mutants, and computational analysis on the structures of IL-10RA proteins were performed. RESULTS We identified a novel compound heterozygote mutation p.[Tyr91Cys];[Pro146Alafs*40] in the IL10RA gene of the patient. The missense variant p.Tyr91Cys was previously identified but not functionally tested, and a frameshift variant, p.Pro146Alafs*40, is novel and nonfunctional. PBMCs from the patient showed defective signal transducer and activator of transcription 3 activation. The p.Tyr91Cys mutant protein failed to properly localize on the plasma membrane. The p.Tyr91Cys mutation seems to disrupt the hydrophobic core structure surrounding the tyrosine 91 residue, causing structural instability. CONCLUSIONS Targeted ES and linkage analysis identified novel compound heterozygous mutations p.[Tyr91Cys];[Pro146Alafs*40] in the IL10RA gene of a child with severe VEO-IBD. p.Tyr91Cys proteins were functionally defective in IL-10RA signaling and failed to properly localize on the plasma membrane, probably due to its structural instability.
Collapse
Affiliation(s)
- Seak Hee Oh
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Hoon Sung
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inki Kim
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chan Kyu Sim
- Lab of Molecular Immunology and Medicine, Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Jung Hoon Lee
- Lab of Molecular Immunology and Medicine, Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Minkyung Baek
- Department of Chemistry, Seoul National University, Seoul, Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chaok Seok
- Department of Chemistry, Seoul National University, Seoul, Korea
| | - Eul Ju Seo
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Myeong Sup Lee
- Lab of Molecular Immunology and Medicine, Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Mo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
20
|
Uhlig HH, Booth C. A Spectrum of Genetic Variants Contributes to Immune Defects and Pathogenesis of Inflammatory Bowel Diseases. Gastroenterology 2018; 154:2022-2024. [PMID: 29733833 DOI: 10.1053/j.gastro.2018.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Holm H Uhlig
- Translational Gastroenterology Unit and Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Claire Booth
- Molecular and Cellular Immunology Section, UCL GOS Institute of Child Health and Department of Paediatric Immunology, Great Ormond Street Hospital, London, UK
| |
Collapse
|
21
|
Disruption of XIAP-RIP2 Association Blocks NOD2-Mediated Inflammatory Signaling. Mol Cell 2018; 69:551-565.e7. [DOI: 10.1016/j.molcel.2018.01.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/26/2017] [Accepted: 01/17/2018] [Indexed: 02/07/2023]
|