Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
2.
J Clin Immunol ; 43(6): 1403-1413, 2023 08.
Article in English | MEDLINE | ID: mdl-37156989

ABSTRACT

PURPOSE: Hyper activation of the JAK-STAT signaling underlies the pathophysiology of many human immune-mediated diseases. Herein, the study of 2 adult patients with SOCS1 haploinsufficiency illustrates the severe and pleomorphic consequences of its impaired regulation in the intestinal tract. METHODS: Two unrelated adult patients presented with gastrointestinal manifestations, one with Crohn's disease-like ileo-colic inflammation refractory to anti-TNF and the other with lymphocytic leiomyositis causing severe chronic intestinal pseudo-occlusion. Next-generation sequencing was used to identify the underlying monogenic defect. One patient received anti-IL-12/IL-23 treatment while the other received the JAK1 inhibitor, ruxolitinib. Peripheral blood, intestinal tissues, and serum samples were analyzed before-and-after JAK1 inhibitor therapy using mass cytometry, histology, transcriptomic, and Olink assay. RESULTS: Novel germline loss-of-function variants in SOCS1 were identified in both patients. The patient with Crohn-like disease achieved clinical remission with anti-IL-12/IL-23 treatment. In the second patient with lymphocytic leiomyositis, ruxolitinib induced rapid resolution of the obstructive symptoms, significant decrease of the CD8+ T lymphocyte muscular infiltrate, and normalization of serum and intestinal cytokines. Decreased frequencies of circulating Treg cells, MAIT cells, and NK cells, with altered CD56bright:CD16lo:CD16hi NK subtype ratios were not modified by ruxolitinib. CONCLUSION: SOCS1 haploinsufficiency can result in a broad spectrum of intestinal manifestations and need to be considered as differential diagnosis in cases of severe treatment-refractory enteropathies, including the rare condition of lymphocytic leiomyositis. This provides the rationale for genetic screening and considering JAK inhibitors in such cases.


Subject(s)
Haploinsufficiency , Tumor Necrosis Factor Inhibitors , Adult , Humans , Suppressor of Cytokine Signaling Proteins/genetics , Interleukin-12 , Interleukin-23 , Suppressor of Cytokine Signaling 1 Protein/genetics
4.
Front Immunol ; 13: 1066375, 2022.
Article in English | MEDLINE | ID: mdl-36569843

ABSTRACT

The transforming growth factor-ß (TGF-ß) family of cytokines exerts pleiotropic functions during embryonic development, tissue homeostasis and repair as well as within the immune system. Single gene defects in individual component of this signaling machinery cause defined Mendelian diseases associated with aberrant activation of TGF-ß signaling, ultimately leading to impaired development, immune responses or both. Gene defects that affect members of the TGF-ß cytokine family result in more restricted phenotypes, while those affecting downstream components of the signaling machinery induce broader defects. These rare disorders, also known as TGF-ß signalopathies, provide the unique opportunity to improve our understanding of the role and the relevance of the TGF-ß signaling in the human immune system. Here, we summarize this elaborate signaling pathway, review the diverse clinical presentations and immunological phenotypes observed in these patients and discuss the phenotypic overlap between humans and mice genetically deficient for individual components of the TGF-ß signaling cascade.


Subject(s)
Loeys-Dietz Syndrome , Transforming Growth Factor beta , Humans , Mice , Animals , Transforming Growth Factor beta/metabolism , Loeys-Dietz Syndrome/genetics , Loeys-Dietz Syndrome/metabolism , Signal Transduction/physiology , Cytokines , Immunity
5.
PLoS One ; 17(10): e0267517, 2022.
Article in English | MEDLINE | ID: mdl-36301921

ABSTRACT

BACKGROUND: Although sepsis is a life-threatening condition, its heterogeneous presentation likely explains the negative results of most trials on adjunctive therapy. This study in patients with sepsis aimed to identify subgroups with similar immune profiles and their clinical and outcome correlates. METHODS: A secondary analysis used data of a prospective multicenter cohort that included patients with early assessment of sepsis. They were described using Predisposition, Insult, Response, Organ failure sepsis (PIRO) staging system. Thirty-eight circulating biomarkers (27 proteins, 11 mRNAs) were assessed at sepsis diagnosis, and their patterns were determined through principal component analysis (PCA). Hierarchical clustering was used to group the patients and k-means algorithm was applied to assess the internal validity of the clusters. RESULTS: Two hundred and three patients were assessed, of median age 64.5 [52.0-77.0] years and SAPS2 score 55 [49-61] points. Five main patterns of biomarkers and six clusters of patients (including 42%, 21%, 17%, 9%, 5% and 5% of the patients) were evidenced. Clusters were distinguished according to the certainty of the causal infection, inflammation, use of organ support, pro- and anti-inflammatory activity, and adaptive profile markers. CONCLUSIONS: In this cohort of patients with suspected sepsis, we individualized clusters which may be described with criteria used to stage sepsis. As these clusters are based on the patterns of circulating biomarkers, whether they might help to predict treatment responsiveness should be addressed in further studies. TRIAL REGISTRATION: The CAPTAIN study was registered on clinicaltrials.gov on June 22, 2011, # NCT01378169.


Subject(s)
Sepsis , Humans , Middle Aged , Prospective Studies , Sepsis/diagnosis , Sepsis/therapy , Biomarkers , Cluster Analysis , Cohort Studies , Intensive Care Units
6.
J Clin Invest ; 132(10)2022 05 16.
Article in English | MEDLINE | ID: mdl-35575086

ABSTRACT

Variants in the UNC45A cochaperone have been recently associated with a syndrome combining diarrhea, cholestasis, deafness, and bone fragility. Yet the mechanism underlying intestinal failure in UNC45A deficiency remains unclear. Here, biallelic variants in UNC45A were identified by next-generation sequencing in 6 patients with congenital diarrhea. Corroborating in silico prediction, variants either abolished UNC45A expression or altered protein conformation. Myosin VB was identified by mass spectrometry as client of the UNC45A chaperone and was found misfolded in UNC45AKO Caco-2 cells. In keeping with impaired myosin VB function, UNC45AKO Caco-2 cells showed abnormal epithelial morphogenesis that was restored by full-length UNC45A, but not by mutant alleles. Patients and UNC45AKO 3D organoids displayed altered luminal development and microvillus inclusions, while 2D cultures revealed Rab11 and apical transporter mislocalization as well as sparse and disorganized microvilli. All those features resembled the subcellular abnormalities observed in duodenal biopsies from patients with microvillus inclusion disease. Finally, microvillus inclusions and shortened microvilli were evidenced in enterocytes from unc45a-deficient zebrafish. Taken together, our results provide evidence that UNC45A plays an essential role in epithelial morphogenesis through its cochaperone function of myosin VB and that UNC45A loss causes a variant of microvillus inclusion disease.


Subject(s)
Diarrhea, Infantile , Malabsorption Syndromes , Mucolipidoses , Myosin Type V , Animals , Caco-2 Cells , Diarrhea, Infantile/metabolism , Diarrhea, Infantile/pathology , Facies , Fetal Growth Retardation , Hair Diseases , Humans , Infant , Intracellular Signaling Peptides and Proteins/metabolism , Malabsorption Syndromes/metabolism , Microvilli/genetics , Microvilli/pathology , Mucolipidoses/genetics , Mucolipidoses/metabolism , Mucolipidoses/pathology , Myosin Type V/genetics , Myosin Type V/metabolism , Phenotype , Zebrafish/genetics , Zebrafish/metabolism
7.
Clin Genet ; 101(5-6): 552-558, 2022 05.
Article in English | MEDLINE | ID: mdl-35132614

ABSTRACT

Variants in aminoacyl-tRNA synthetases (ARSs) genes are associated to a broad spectrum of human inherited diseases. Patients with defective PheRS, encoded by FARSA and FARSB, display brain abnormalities, interstitial lung disease and facial dysmorphism. We investigated four children from two unrelated consanguineous families carrying two missense homozygous variants in FARSA with significantly reduced PheRS-mediated aminoacylation activity. In addition to the core ARS-phenotype, all patients showed an inflammatory profile associated with autoimmunity and interferon score, a clinical feature not ascribed to PheRS-deficient patients to date. JAK inhibition improved lung disease in one patient. Our findings expand the genetic and clinical spectrum of FARSA-related disease.


Subject(s)
Amino Acyl-tRNA Synthetases , Charcot-Marie-Tooth Disease , Lung Diseases, Interstitial , Amino Acyl-tRNA Synthetases/genetics , Charcot-Marie-Tooth Disease/genetics , Consanguinity , Humans , Lung Diseases, Interstitial/genetics , Phenotype , Syndrome
8.
Protein Sci ; 31(2): 538-544, 2022 02.
Article in English | MEDLINE | ID: mdl-34806245

ABSTRACT

PTPN2 is an important protein tyrosine phosphatase (PTP) that plays a key role in cell signaling. Deletions or inactivating mutations of PTPN2 have been described in different pathologies and underline its critical role in hematopoiesis, autoimmunity, and inflammation. Surprisingly, despite the major pathophysiological implications of PTPN2, the structural analysis of this PTP and notably of its pathogenic mutants remains poorly documented. Contrary to other human PTP enzymes, to date, only one structure of PTPN2 (wild-type form) has been reported. Here, we report the first crystal structure of a pathogenic mutant of PTPN2 (Cys216Gly) that causes an autoimmune enteropathy. We show in particular that this mutant adopts a classical PTP fold. More importantly, albeit inactive, the mutant retains its ability to bind substrates and to adopt the characteristic catalytically competent closed form of PTP enzymes. This novel PTPN2 structure may serve as a new tool to better understand PTP structures and the structural impacts of pathogenic mutations. Moreover, the C216G PTPN2 structure could also be helpful to design specific ligands/inhibitors.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Signal Transduction , Humans , Polyendocrinopathies, Autoimmune/genetics , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism
9.
Mucosal Immunol ; 14(5): 1172-1182, 2021 09.
Article in English | MEDLINE | ID: mdl-34226674

ABSTRACT

Single genetic mutations predispose to very early onset inflammatory bowel disease (VEO-IBD). Here, we identify a de novo duplication of the 10p15.1 chromosomal region, including the IL2RA locus, in a 2-year-old girl with treatment-resistant pancolitis that was brought into remission by colectomy. Strikingly, after colectomy while the patient was in clinical remission and without medication, the peripheral blood CD4:CD8 ratio was constitutively high and CD25 expression was increased on circulating effector memory, Foxp3+, and Foxp3neg CD4+ T cells compared to healthy controls. This high CD25 expression increased IL-2 signaling, potentiating CD4+ T-cell-derived IFNγ secretion after T-cell receptor (TCR) stimulation. Restoring CD25 expression using the JAK1/3-inhibitor tofacitinib controlled TCR-induced IFNγ secretion in vitro. As diseased colonic tissue, but not the unaffected duodenum, contained mainly CD4+ T cells with a prominent IFNγ-signature, we hypothesize that local microbial stimulation may have initiated colonic disease. Overall, we identify that duplication of the IL2RA locus can associate with VEO-IBD and suggest that increased IL-2 signaling predisposes to colonic intestinal inflammation.


Subject(s)
Colitis/etiology , Colitis/metabolism , Gene Duplication , Genetic Predisposition to Disease , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2/metabolism , Signal Transduction , Age of Onset , Alleles , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Chromosomes, Human, Pair 10 , Colitis/diagnosis , Cytokines/metabolism , Drug Resistance , Gene Expression , Genetic Association Studies , Genetic Loci , Humans , Immunohistochemistry , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
10.
Am J Hum Genet ; 108(6): 1126-1137, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34010604

ABSTRACT

Dysregulated transforming growth factor TGF-ß signaling underlies the pathogenesis of genetic disorders affecting the connective tissue such as Loeys-Dietz syndrome. Here, we report 12 individuals with bi-allelic loss-of-function variants in IPO8 who presented with a syndromic association characterized by cardio-vascular anomalies, joint hyperlaxity, and various degree of dysmorphic features and developmental delay as well as immune dysregulation; the individuals were from nine unrelated families. Importin 8 belongs to the karyopherin family of nuclear transport receptors and was previously shown to mediate TGF-ß-dependent SMADs trafficking to the nucleus in vitro. The important in vivo role of IPO8 in pSMAD nuclear translocation was demonstrated by CRISPR/Cas9-mediated inactivation in zebrafish. Consistent with IPO8's role in BMP/TGF-ß signaling, ipo8-/- zebrafish presented mild to severe dorso-ventral patterning defects during early embryonic development. Moreover, ipo8-/- zebrafish displayed severe cardiovascular and skeletal defects that mirrored the human phenotype. Our work thus provides evidence that IPO8 plays a critical and non-redundant role in TGF-ß signaling during development and reinforces the existing link between TGF-ß signaling and connective tissue defects.


Subject(s)
Bone Diseases/etiology , Cardiovascular Diseases/etiology , Connective Tissue Diseases/etiology , Immunity, Cellular/immunology , Loss of Function Mutation , Loss of Heterozygosity , beta Karyopherins/genetics , Adolescent , Adult , Animals , Bone Diseases/pathology , Cardiovascular Diseases/pathology , Child , Connective Tissue Diseases/pathology , Female , Humans , Infant , Male , Middle Aged , Pedigree , Phenotype , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Young Adult , Zebrafish , beta Karyopherins/metabolism
11.
Mucosal Immunol ; 14(5): 1017-1037, 2021 09.
Article in English | MEDLINE | ID: mdl-33859369

ABSTRACT

The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.


Subject(s)
Host Microbial Interactions/immunology , Immunity, Mucosal , Immunomodulation , Intestinal Mucosa/physiology , Adaptive Immunity , Animals , Biological Transport , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Disease Susceptibility , Enterocytes/cytology , Enterocytes/immunology , Enterocytes/metabolism , Gene Expression Regulation , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/immunology , Genetic Predisposition to Disease , Hematopoiesis/genetics , Hematopoiesis/immunology , Homeostasis , Host Microbial Interactions/genetics , Humans , Immunity, Innate , Mutation , Signal Transduction
16.
Intensive Care Med ; 44(7): 1061-1070, 2018 07.
Article in English | MEDLINE | ID: mdl-29959455

ABSTRACT

PURPOSE: Sepsis and non-septic systemic inflammatory response syndrome (SIRS) are the same syndromes, differing by their cause, sepsis being secondary to microbial infection. Microbiological tests are not enough to detect infection early. While more than 50 biomarkers have been proposed to detect infection, none have been repeatedly validated. AIM: To assess the accuracy of circulating biomarkers to discriminate between sepsis and non-septic SIRS. METHODS: The CAPTAIN study was a prospective observational multicenter cohort of 279 ICU patients with hypo- or hyperthermia and criteria of SIRS, included at the time the attending physician considered antimicrobial therapy. Investigators collected blood at inclusion to measure 29 plasma compounds and ten whole blood RNAs, and-for those patients included within working hours-14 leukocyte surface markers. Patients were classified as having sepsis or non-septic SIRS blindly to the biomarkers results. We used the LASSO method as the technique of multivariate analysis, because of the large number of biomarkers. RESULTS: During the study period, 363 patients with SIRS were screened, 84 having exclusion criteria. Ninety-one patients were classified as having non-septic SIRS and 188 as having sepsis. Eight biomarkers had an area under the receiver operating curve (ROC-AUC) over 0.6 with a 95% confidence interval over 0.5. LASSO regression identified CRP and HLA-DRA mRNA as being repeatedly associated with sepsis, and no model performed better than CRP alone (ROC-AUC 0.76 [0.68-0.84]). CONCLUSIONS: The circulating biomarkers tested were found to discriminate poorly between sepsis and non-septic SIRS, and no combination performed better than CRP alone.


Subject(s)
Biomarkers , Sepsis , Systemic Inflammatory Response Syndrome , Aged , Biomarkers/blood , Diagnosis, Differential , Female , Humans , Intensive Care Units , Male , Middle Aged , Prospective Studies , Sepsis/blood , Sepsis/diagnosis , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/diagnosis
17.
J Crohns Colitis ; 12(9): 1104-1112, 2018 08 29.
Article in English | MEDLINE | ID: mdl-29788237

ABSTRACT

BACKGROUND AND AIMS: An expanding number of monogenic defects have been identified as causative of severe forms of very early-onset inflammatory bowel diseases [VEO-IBD]. The present study aimed at defining how next-generation sequencing [NGS] methods can be used to improve identification of known molecular diagnosis and to adapt treatment. METHODS: A total of 207 children were recruited in 45 paediatric centres through an international collaborative network [ESPGHAN GENIUS working group] with a clinical presentation of severe VEO-IBD [n = 185] or an anamnesis suggestive of a monogenic disorder [n = 22]. Patients were divided at inclusion into three phenotypic subsets: predominantly small bowel inflammation, colitis with perianal lesions, and colitis only. Methods to obtain molecular diagnosis included functional tests followed by specific Sanger sequencing, custom-made targeted NGS, and in selected cases whole exome sequencing [WES] of parents-child trios. Genetic findings were validated clinically and/or functionally. RESULTS: Molecular diagnosis was achieved in 66/207 children [32%]: 61% with small bowel inflammation, 39% with colitis and perianal lesions, and 18% with colitis only. Targeted NGS pinpointed gene mutations causative of atypical presentations, and identified large exonic copy number variations previously missed by WES. CONCLUSIONS: Our results lead us to propose an optimised diagnostic strategy to identify known monogenic causes of severe IBD.


Subject(s)
High-Throughput Nucleotide Sequencing , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/etiology , Adolescent , Age of Onset , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Inflammatory Bowel Diseases/therapy , Male , Predictive Value of Tests
18.
EMBO Mol Med ; 10(4)2018 04.
Article in English | MEDLINE | ID: mdl-29567797

ABSTRACT

Herein, we report the first identification of biallelic-inherited mutations in ALPI as a Mendelian cause of inflammatory bowel disease in two unrelated patients. ALPI encodes for intestinal phosphatase alkaline, a brush border metalloenzyme that hydrolyses phosphate from the lipid A moiety of lipopolysaccharides and thereby drastically reduces Toll-like receptor 4 agonist activity. Prediction tools and structural modelling indicate that all mutations affect critical residues or inter-subunit interactions, and heterologous expression in HEK293T cells demonstrated that all ALPI mutations were loss of function. ALPI mutations impaired either stability or catalytic activity of ALPI and rendered it unable to detoxify lipopolysaccharide-dependent signalling. Furthermore, ALPI expression was reduced in patients' biopsies, and ALPI activity was undetectable in ALPI-deficient patient's stool. Our findings highlight the crucial role of ALPI in regulating host-microbiota interactions and restraining host inflammatory responses. These results indicate that ALPI mutations should be included in screening for monogenic causes of inflammatory bowel diseases and lay the groundwork for ALPI-based treatments in intestinal inflammatory disorders.


Subject(s)
Alkaline Phosphatase/metabolism , Inflammatory Bowel Diseases/metabolism , Alkaline Phosphatase/deficiency , Alkaline Phosphatase/genetics , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , Homeostasis , Humans , Inflammatory Bowel Diseases/genetics , Intestines , Mutation/genetics , Signal Transduction/physiology
20.
J Pediatr Gastroenterol Nutr ; 64(3): 378-384, 2017 03.
Article in English | MEDLINE | ID: mdl-27253662

ABSTRACT

OBJECTIVE: Early-onset inflammatory bowel diseases can result from a wide spectrum of rare mendelian disorders. Early molecular diagnosis is crucial in defining treatment and in improving life expectancy. Herein we aimed at defining the mechanism of an immunodeficiency-polyendrocrinopathy and enteropathy-X-linked (IPEX)-like disease combined with a severe immunodeficiency in 2 siblings born from distantly related parents. METHODS: Whole exome sequencing was performed on blood-extracted genomic DNA from the 2 affected children and their parents on the genomic platform of Institut IMAGINE. Candidate gene mutation was identified using the in-house software PolyWeb and confirmed by Sanger sequencing. Protein expression was determined by western blot. Flow cytometry was used to assess consequences of the mutation on lymphocyte phenotype and nuclear factor-kappa B (NF-κB) activation at diagnosis and after treatment by hematopoietic stem cell transplantation. RESULTS: We identified a homozygous missense mutation in mucosa-associated lymphoid tissue lymphoma translocation 1 gene (MALT1), which precluded protein expression. In keeping with the known function of MALT1, NF-κB-dependent lymphocyte activation was severely impaired. Moreover, there was a drastic reduction in Forkhead box P3 (FOXP3) regulatory T cells accounting for the IPEX-like phenotype. Following identification of the mutation, both children received hematopoietic stem cell transplantation, which permitted full clinical recovery. Immunological workup at 6 and 12 months after transplantation showed normal NF-κB activation and correction of regulatory T cells frequency. CONCLUSIONS: Along with FOXP3, interleukin 2 receptor alpha chain (IL2RA), and cytotoxic T-lymphocyte protein 4 precursor (CTLA-4) mutations, MALT1 deficiency should now be considered as a possible cause of IPEX-like syndrome associated with immunodeficiency that can be cured by hematopoietic stem cell transplantation.


Subject(s)
Diabetes Mellitus, Type 1/congenital , Diarrhea/genetics , Genetic Diseases, X-Linked/genetics , Immune System Diseases/congenital , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/deficiency , Mutation, Missense , Child , Child, Preschool , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/genetics , Diarrhea/diagnosis , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Markers , Homozygote , Humans , Immune System Diseases/diagnosis , Immune System Diseases/genetics , Male , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Siblings
SELECTION OF CITATIONS
SEARCH DETAIL
...