Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Sci Adv ; 9(35): eadg6375, 2023 09.
Article in English | MEDLINE | ID: mdl-37656781

ABSTRACT

Although research on rare autoimmune and autoinflammatory diseases has enabled definition of nonredundant regulators of homeostasis in human immunity, because of the single gene-single disease nature of many of these diseases, contributing factors were mostly unveiled in sequential and noncoordinated individual studies. We used a network-based approach for integrating a set of 186 inborn errors of immunity with predominant autoimmunity/autoinflammation into a comprehensive map of human immune dysregulation, which we termed "AutoCore." The AutoCore is located centrally within the interactome of all protein-protein interactions, connecting and pinpointing multidisease markers for a range of common, polygenic autoimmune/autoinflammatory diseases. The AutoCore can be subdivided into 19 endotypes that correspond to molecularly and phenotypically cohesive disease subgroups, providing a molecular mechanism-based disease classification and rationale toward systematic targeting for therapeutic purposes. Our study provides a proof of concept for using network-based methods to systematically investigate the molecular relationships between individual rare diseases and address a range of conceptual, diagnostic, and therapeutic challenges.


Subject(s)
Autoimmune Diseases , Hereditary Autoinflammatory Diseases , Humans , Autoimmunity , Homeostasis
2.
N Engl J Med ; 389(6): 527-539, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37342957

ABSTRACT

BACKGROUND: Increasing evidence links genetic defects affecting actin-regulatory proteins to diseases with severe autoimmunity and autoinflammation, yet the underlying molecular mechanisms are poorly understood. Dedicator of cytokinesis 11 (DOCK11) activates the small Rho guanosine triphosphatase (GTPase) cell division cycle 42 (CDC42), a central regulator of actin cytoskeleton dynamics. The role of DOCK11 in human immune-cell function and disease remains unknown. METHODS: We conducted genetic, immunologic, and molecular assays in four patients from four unrelated families who presented with infections, early-onset severe immune dysregulation, normocytic anemia of variable severity associated with anisopoikilocytosis, and developmental delay. Functional assays were performed in patient-derived cells, as well as in mouse and zebrafish models. RESULTS: We identified rare, X-linked germline mutations in DOCK11 in the patients, leading to a loss of protein expression in two patients and impaired CDC42 activation in all four patients. Patient-derived T cells did not form filopodia and showed abnormal migration. In addition, the patient-derived T cells, as well as the T cells from Dock11-knockout mice, showed overt activation and production of proinflammatory cytokines that were associated with an increased degree of nuclear translocation of nuclear factor of activated T cell 1 (NFATc1). Anemia and aberrant erythrocyte morphologic features were recapitulated in a newly generated dock11-knockout zebrafish model, and anemia was amenable to rescue on ectopic expression of constitutively active CDC42. CONCLUSIONS: Germline hemizygous loss-of-function mutations affecting the actin regulator DOCK11 were shown to cause a previously unknown inborn error of hematopoiesis and immunity characterized by severe immune dysregulation and systemic inflammation, recurrent infections, and anemia. (Funded by the European Research Council and others.).


Subject(s)
Actins , Anemia , Guanine Nucleotide Exchange Factors , Inflammation , Animals , Humans , Mice , Actins/genetics , Actins/metabolism , Anemia/etiology , Anemia/genetics , Disease Models, Animal , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Hematopoiesis , Inflammation/etiology , Inflammation/genetics , Zebrafish/genetics , Zebrafish/metabolism
3.
Blood ; 142(9): 827-845, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37249233

ABSTRACT

The nuclear factor of activated T cells (NFAT) family of transcription factors plays central roles in adaptive immunity in murine models; however, their contribution to human immune homeostasis remains poorly defined. In a multigenerational pedigree, we identified 3 patients who carry germ line biallelic missense variants in NFATC1, presenting with recurrent infections, hypogammaglobulinemia, and decreased antibody responses. The compound heterozygous NFATC1 variants identified in these patients caused decreased stability and reduced the binding of DNA and interacting proteins. We observed defects in early activation and proliferation of T and B cells from these patients, amenable to rescue upon genetic reconstitution. Stimulation induced early T-cell activation and proliferation responses were delayed but not lost, reaching that of healthy controls at day 7, indicative of an adaptive capacity of the cells. Assessment of the metabolic capacity of patient T cells revealed that NFATc1 dysfunction rendered T cells unable to engage in glycolysis after stimulation, although oxidative metabolic processes were intact. We hypothesized that NFATc1-mutant T cells could compensate for the energy deficit due to defective glycolysis by using enhanced lipid metabolism as an adaptation, leading to a delayed, but not lost, activation responses. Indeed, we observed increased 13C-labeled palmitate incorporation into citrate, indicating higher fatty acid oxidation, and we demonstrated that metformin and rosiglitazone improved patient T-cell effector functions. Collectively, enabled by our molecular dissection of the consequences of loss-of-function NFATC1 mutations and extending the role of NFATc1 in human immunity beyond receptor signaling, we provide evidence of metabolic plasticity in the context of impaired glycolysis observed in patient T cells, alleviating delayed effector responses.


Subject(s)
NFATC Transcription Factors , T-Lymphocytes , Humans , Mice , Animals , T-Lymphocytes/metabolism , NFATC Transcription Factors/metabolism , CD8-Positive T-Lymphocytes , Glycolysis/genetics , Mutation
5.
Turk J Haematol ; 37(4): 271-281, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32812413

ABSTRACT

Objective: Wiskott-Aldrich syndrome (WAS) is an X-linked primary immune deficiency characterized by microthrombocytopenia, eczema, and recurrent infections. We aimed to evaluate the clinical features and outcomes of a WAS cohort. Materials and Methods: We retrospectively evaluated the clinical courses, immunological features, treatments, and outcomes in a total of 23 WAS patients together with data related to 11 transplanted cases among them between 1982 and 2019. Results: Before admission, 11 patients (48%) were misdiagnosed with immune thrombocytopenia. WAS scores were mostly 4 or 5. Eleven patients were transplanted and they had an overall survival rate of 100% during a median follow-up period of 8.5 years (range: 8 months to 20 years). Five patients who were not transplanted died at a median of 7 years (range: 2-26 years). Nontransplanted patients had high morbidity due to organ damage, mostly caused by autoimmunity, bleeding, and infections. Two novel mutations were also defined. Conclusion: All male babies with microthrombocytopenia should be evaluated for WAS. Hematopoietic stem cell transplantation should be performed at the earliest age with the best possible donors.


Subject(s)
Phenotype , Wiskott-Aldrich Syndrome/diagnosis , Adolescent , Biomarkers , Child , Child, Preschool , Combined Modality Therapy , Diagnosis, Differential , Disease Management , Disease Susceptibility , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunoglobulins, Intravenous/therapeutic use , Infant , Infant, Newborn , Male , Patient Outcome Assessment , Prognosis , Reinfection/etiology , Symptom Assessment , Treatment Outcome , Wiskott-Aldrich Syndrome/complications , Wiskott-Aldrich Syndrome/etiology , Wiskott-Aldrich Syndrome/therapy , Young Adult
6.
Blood ; 136(23): 2638-2655, 2020 12 03.
Article in English | MEDLINE | ID: mdl-32603431

ABSTRACT

Biallelic mutations in the genes encoding CD27 or its ligand CD70 underlie inborn errors of immunity (IEIs) characterized predominantly by Epstein-Barr virus (EBV)-associated immune dysregulation, such as chronic viremia, severe infectious mononucleosis, hemophagocytic lymphohistiocytosis (HLH), lymphoproliferation, and malignancy. A comprehensive understanding of the natural history, immune characteristics, and transplant outcomes has remained elusive. Here, in a multi-institutional global collaboration, we collected the clinical information of 49 patients from 29 families (CD27, n = 33; CD70, n = 16), including 24 previously unreported individuals and identified a total of 16 distinct mutations in CD27, and 8 in CD70, respectively. The majority of patients (90%) were EBV+ at diagnosis, but only ∼30% presented with infectious mononucleosis. Lymphoproliferation and lymphoma were the main clinical manifestations (70% and 43%, respectively), and 9 of the CD27-deficient patients developed HLH. Twenty-one patients (43%) developed autoinflammatory features including uveitis, arthritis, and periodic fever. Detailed immunological characterization revealed aberrant generation of memory B and T cells, including a paucity of EBV-specific T cells, and impaired effector function of CD8+ T cells, thereby providing mechanistic insight into cellular defects underpinning the clinical features of disrupted CD27/CD70 signaling. Nineteen patients underwent allogeneic hematopoietic stem cell transplantation (HSCT) prior to adulthood predominantly because of lymphoma, with 95% survival without disease recurrence. Our data highlight the marked predisposition to lymphoma of both CD27- and CD70-deficient patients. The excellent outcome after HSCT supports the timely implementation of this treatment modality particularly in patients presenting with malignant transformation to lymphoma.


Subject(s)
CD27 Ligand/deficiency , Genetic Diseases, Inborn , Hematopoietic Stem Cell Transplantation , Immunologic Deficiency Syndromes , Tumor Necrosis Factor Receptor Superfamily, Member 7/deficiency , Adolescent , Adult , Allografts , Child , Child, Preschool , Disease-Free Survival , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/immunology , Genetic Diseases, Inborn/mortality , Genetic Diseases, Inborn/therapy , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/mortality , Immunologic Deficiency Syndromes/therapy , Infant , Male , Retrospective Studies , Survival Rate
7.
Pediatr Allergy Immunol ; 31(5): 515-527, 2020 07.
Article in English | MEDLINE | ID: mdl-32108967

ABSTRACT

Biallelic mutations in the dedicator of cytokinesis 8 gene (DOCK8) cause a progressive combined immunodeficiency (CID) characterized by susceptibility to severe viral skin infections, atopic diseases, recurrent respiratory infections, and malignancy. Hematopoietic stem cell transplantation (HSCT) is only curative treatment for the disease. However, there is limited information about long-term outcome of HSCT and its effect to protect against cancer development in DOCK8-deficient patients. In this study, we retrospectively evaluated clinical and immunologic characteristics of 20 DOCK8-deficient patients and outcome of 11 patients who underwent HSCT. We aimed to report the experience of our center and the result of the largest transplantation series of DOCK8 deficiency in our country. Median follow-up time is 71 months (min-max: 16-172) in all patients and 48 months (min-max: 5-84) in transplanted patients. Atopic dermatitis (18/20), recurrent respiratory tract infections (17/20), and food allergy (14/20) were the most frequent clinical manifestations. Failure to thrive (13/20), liver problems (12/20), bronchiectasis (11/20), chronic diarrhea (10/21), and autism spectrum disorders (3/20) were remarkable findings in our series. Elevated IgE level (20/20) and eosinophilia (17/20), low IgM level (15/20), and decreased CD3+ T (10/20) and CD4+ T (11/20) cell count were prominent laboratory findings. HSCT was performed in 11 patients. All patients achieved adequate engraftment and showed improvement in their clinical and immunologic findings. Atopic dermatitis and food allergies improved in all patients, and their dietary restriction was stopped except one patient who was transplanted recently. The frequency of infections was decreased. The overall survival is 91% in HSCT-received patients and 80% in all. HSCT at the earliest possible period with most suitable donor- and patient-specific appropriate conditioning regimen and GvHD prophylaxis is lifesaving for DOCK8 deficiency cases.


Subject(s)
Guanine Nucleotide Exchange Factors , Hematopoietic Stem Cell Transplantation , Cytokinesis , Follow-Up Studies , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Homozygote , Humans , Infant, Newborn , Retrospective Studies , Sequence Deletion
8.
J Allergy Clin Immunol ; 145(5): 1452-1463, 2020 05.
Article in English | MEDLINE | ID: mdl-31887391

ABSTRACT

BACKGROUND: Recent findings strongly support hematopoietic stem cell transplantation (HSCT) in patients with severe presentation of LPS-responsive beige-like anchor protein (LRBA) deficiency, but long-term follow-up and survival data beyond previous patient reports or meta-reviews are scarce for those patients who do not receive a transplant. OBJECTIVE: This international retrospective study was conducted to elucidate the longitudinal clinical course of patients with LRBA deficiency who do and do not receive a transplant. METHOD: We assessed disease burden and treatment responses with a specially developed immune deficiency and dysregulation activity score, reflecting the sum and severity of organ involvement and infections, days of hospitalization, supportive care requirements, and performance indices. RESULTS: Of 76 patients with LRBA deficiency from 29 centers (median follow-up, 10 years; range, 1-52), 24 underwent HSCT from 2005 to 2019. The overall survival rate after HSCT (median follow-up, 20 months) was 70.8% (17 of 24 patients); all deaths were due to nonspecific, early, transplant-related mortality. Currently, 82.7% of patients who did not receive a transplant (43 of 52; age range, 3-69 years) are alive. Of 17 HSCT survivors, 7 are in complete remission and 5 are in good partial remission without treatment (together, 12 of 17 [70.6%]). In contrast, only 5 of 43 patients who did not receive a transplant (11.6%) are without immunosuppression. Immune deficiency and dysregulation activity scores were significantly lower in patients who survived HSCT than in those receiving conventional treatment (P = .005) or in patients who received abatacept or sirolimus as compared with other therapies, and in patients with residual LRBA expression. Higher disease burden, longer duration before HSCT, and lung involvement were associated with poor outcome. CONCLUSION: The lifelong disease activity, implying a need for immunosuppression and risk of malignancy, must be weighed against the risks of HSCT.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Hematopoietic Stem Cell Transplantation , Immunologic Deficiency Syndromes/therapy , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Immunologic Deficiency Syndromes/mortality , Male , Middle Aged , Survival Analysis , Treatment Outcome , Young Adult
9.
Pediatr Res ; 87(2): 293-299, 2020 01.
Article in English | MEDLINE | ID: mdl-31581173

ABSTRACT

Rare monogenetic diseases serve as natural models to dissect the molecular pathophysiology of the complex disease traits. Rheumatologic disorders by their nature are considered complex diseases with partially genetic origin, as illustrated by their heterogeneous genetic background and variable phenotypic presentation. Recent advances in genetic technologies have helped uncover multiple variants associated with disease susceptibility; however, a precise understanding of genotype-phenotype relationships is still missing. Inborn errors of immunity (IEIs), in addition to recurrent infections, may also present with autoimmune and autoinflammatory rheumatologic manifestations and have provided insights for understanding the underlying the principles of immune system homeostasis and mechanisms of immune dysregulation. This review discusses the rheumatologic manifestations in IEIs with overlapping and differentiating features in immunodeficiencies and rheumatologic disorders.


Subject(s)
Autoimmunity , Immune System/immunology , Primary Immunodeficiency Diseases/immunology , Rheumatic Diseases/immunology , Animals , Autoimmunity/genetics , Genetic Predisposition to Disease , Humans , Immune System/growth & development , Phenotype , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/physiopathology , Rheumatic Diseases/genetics , Rheumatic Diseases/physiopathology , Risk Assessment , Risk Factors
10.
J Exp Med ; 216(9): 1986-1998, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31235509

ABSTRACT

IL-6 excess is central to the pathogenesis of multiple inflammatory conditions and is targeted in clinical practice by immunotherapy that blocks the IL-6 receptor encoded by IL6R We describe two patients with homozygous mutations in IL6R who presented with recurrent infections, abnormal acute-phase responses, elevated IgE, eczema, and eosinophilia. This study identifies a novel primary immunodeficiency, clarifying the contribution of IL-6 to the phenotype of patients with mutations in IL6ST, STAT3, and ZNF341, genes encoding different components of the IL-6 signaling pathway, and alerts us to the potential toxicity of drugs targeting the IL-6R.


Subject(s)
Immunologic Deficiency Syndromes/pathology , Inflammation/pathology , Receptors, Interleukin-6/deficiency , Adolescent , Adult , Child , Child, Preschool , Female , HEK293 Cells , Humans , Infant, Newborn , Male , Receptors, Interleukin-6/metabolism
11.
Pediatr Transplant ; 22(7): e13266, 2018 11.
Article in English | MEDLINE | ID: mdl-29992714

ABSTRACT

INTRODUCTION: HSCT is the curative therapeutic option in PIDs. Due to the increase in survival rates, reduced-toxicity conditioning regimens with treosulfan have become another alternative. The purpose of this retrospective study was to analyze the outcome of treosulfan-based conditioning before HSCT for patients with PID. METHOD: A total of 15 patients that received a treosulfan-based conditioning regimen for HSCT were recruited. Type of diagnosis, donor and stem cell source, pretransplant organ damage, infections, engraftment, chimerism, and transplant-related toxicities were analyzed. RESULTS: At a median follow-up time of 32 months, the overall survival was 86.7%. Following HSCT, 14 of 15 patients had engraftment, with 86.7% of the cohort having full-donor chimerism. The most common toxicity was seen on the skin (53.3%). Acute GVHD and chronic GVHD were documented in 53% and 20% of the study population, respectively. Although the cohort consisted of patients with pretransplant liver damage, SOS manifestations were documented in 20%. CONCLUSION: Treosulfan-based conditioning regimens before HSCT are associated with lower toxicity compared to myeloablative regimens, are safe, and have high engraftment rates with full-donor chimerism in patients having PID, regardless of the specified genetic diagnosis and donor type.


Subject(s)
Busulfan/analogs & derivatives , Hematopoietic Stem Cell Transplantation , Immunologic Deficiency Syndromes/therapy , Immunosuppressive Agents/administration & dosage , Transplantation Conditioning/methods , Adolescent , Busulfan/administration & dosage , Busulfan/therapeutic use , Child , Child, Preschool , Drug Administration Schedule , Drug Therapy, Combination , Female , Follow-Up Studies , Graft vs Host Disease/prevention & control , Humans , Immunologic Deficiency Syndromes/mortality , Immunosuppressive Agents/therapeutic use , Infant , Male , Retrospective Studies , Survival Analysis , Treatment Outcome
12.
J Clin Immunol ; 37(8): 790-800, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28956255

ABSTRACT

INTRODUCTION: LPS-responsive beige-like anchor protein (LRBA) deficiency is a primary immunodeficiency categorized as common variable immunodeficiency associated with autoimmune manifestations and inflammatory bowel diseases; however, the clinical spectrum has been extended. Here, we present our cohort of Turkish LRBA-deficient patients from a single center, demonstrating a diversity of clinical manifestations. METHOD: Seven affected individuals from five families were assessed retrospectively in this study. RESULTS: Of the seven patients with LRBA deficiency, four had homozygous, and two had compound heterozygous mutations. One patient remained disease free until the last follow-up (age 17 years). The most common clinical manifestations of the six symptomatic patients were organomegaly (6/6), autoimmunity (6/6), and chronic diarrhea (5/6). Recurrent infectious episodes were observed in three patients. None of the patients had hypogammaglobulinemia at presentation. B cell subpopulation analysis revealed low numbers of switched-memory B cell numbers in two of the four tested patients. During the disease course, three of the patients died, two of them underwent successful hematopoietic stem cell transplantation (HSCT) from matched sibling donors, and one is under abatacept therapy. CONCLUSION: LRBA defects should always be kept in mind as a differential diagnosis for patients with autoimmune disease affecting multiple organs, chronic diarrhea, and organomegalies. In our experience, early HSCT is a life-saving therapeutic strategy.


Subject(s)
Abatacept/therapeutic use , Adaptor Proteins, Signal Transducing/genetics , Autoimmune Diseases/genetics , Common Variable Immunodeficiency/genetics , Immunosuppressive Agents/therapeutic use , Inflammatory Bowel Diseases/genetics , Mutation/genetics , Adolescent , Autoimmune Diseases/diagnosis , Autoimmune Diseases/drug therapy , Child , Common Variable Immunodeficiency/diagnosis , Common Variable Immunodeficiency/drug therapy , Disease-Free Survival , Fatal Outcome , Female , Homozygote , Humans , Infant , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/drug therapy , Male , Sepsis , Turkey
SELECTION OF CITATIONS
SEARCH DETAIL
...