Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JCEM Case Rep ; 2(6): luae091, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38832003

RESUMEN

Rapid onset obesity with hypoventilation, hypothalamic, and autonomic dysregulation (ROHHAD) syndrome in childhood is characterized by abrupt onset weight gain and dysautonomia with variable neuroendocrine involvement. In the absence of definitive disease-modifying therapies, the primary management strategy remains symptom control. This case report describes the first successful correction of obesity, dysautonomia, and metabolic derangement in a patient with ROHHAD following Roux-en-Y gastric bypass. Anthropometrics, metabolic profiling, and stool microbiome composition were assessed in a longitudinal fashion. In the 48-month period following surgery, the patient body mass index (BMI) reduced by 9.5 kg/m2 and metabolic status improved, evidenced in weaning of insulin, and improved glycated hemoglobin, lipid profile, and hepatic enzymes. Chronic diarrhea resolved after surgery and prior to significant weight loss. Evaluation of stool bacterial composition and biomass demonstrated shifts in absolute abundance and taxonomic composition in longitudinal samples following surgery. This case demonstrates the potential efficacy of bariatric surgery in correcting the metabolic disruption of ROHHAD syndrome, producing long-term changes in gut microbiome composition and biomass.

2.
Sci Transl Med ; 15(719): eadh0353, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37878676

RESUMEN

Immune-targeted therapies have efficacy for treatment of autoinflammatory diseases. For example, treatment with the T cell-specific anti-CD3 antibody teplizumab delayed disease onset in participants at high risk for type 1 diabetes (T1D) in the TrialNet 10 (TN-10) trial. However, heterogeneity in therapeutic responses in TN-10 and other immunotherapy trials identifies gaps in understanding disease progression and treatment responses. The intestinal microbiome is a potential source of biomarkers associated with future T1D diagnosis and responses to immunotherapy. We previously reported that antibody responses to gut commensal bacteria were associated with T1D diagnosis, suggesting that certain antimicrobial immune responses may help predict disease onset. Here, we investigated anticommensal antibody (ACAb) responses against a panel of taxonomically diverse intestinal bacteria species in sera from TN-10 participants before and after teplizumab or placebo treatment. We identified IgG2 responses to three species that were associated with time to T1D diagnosis and with teplizumab treatment responses that delayed disease onset. These antibody responses link human intestinal bacteria with T1D progression, adding predictive value to known T1D risk factors. ACAb analysis provides a new approach to elucidate heterogeneity in responses to immunotherapy and identify individuals who may benefit from teplizumab, recently approved by the U.S. Food and Drug Administration for delaying T1D onset.


Asunto(s)
Diabetes Mellitus Tipo 1 , Humanos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Inmunoterapia , Linfocitos T , Bacterias , Inmunidad
3.
Metabolism ; 149: 155695, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37802200

RESUMEN

BACKGROUND: Gestational diabetes (GDM) is a distinctive form of diabetes that first presents in pregnancy. While most women return to normoglycemia after delivery, they are nearly ten times more likely to develop type 2 diabetes than women with uncomplicated pregnancies. Current prevention strategies remain limited due to our incomplete understanding of the early underpinnings of progression. AIM: To comprehensively characterize the postpartum profiles of women shortly after a GDM pregnancy and identify key mechanisms responsible for the progression to overt type 2 diabetes using multi-dimensional approaches. METHODS: We conducted a nested case-control study of 200 women from the Study of Women, Infant Feeding and Type 2 Diabetes After GDM Pregnancy (SWIFT) to examine biochemical, proteomic, metabolomic, and lipidomic profiles at 6-9 weeks postpartum (baseline) after a GDM pregnancy. At baseline and annually up to two years, SWIFT administered research 2-hour 75-gram oral glucose tolerance tests. Women who developed incident type 2 diabetes within four years of delivery (incident case group, n = 100) were pair-matched by age, race, and pre-pregnancy body mass index to those who remained free of diabetes for at least 8 years (control group, n = 100). Correlation analyses were used to assess and integrate relationships across profiling platforms. RESULTS: At baseline, all 200 women were free of diabetes. The case group was more likely to present with dysglycemia (e.g., impaired fasting glucose levels, glucose tolerance, or both). We also detected differences between groups across all omic platforms. Notably, protein profiles revealed an underlying inflammatory response with perturbations in protease inhibitors, coagulation components, extracellular matrix components, and lipoproteins, whereas metabolite and lipid profiles implicated disturbances in amino acids and triglycerides at individual and class levels with future progression. We identified significant correlations between profile features and fasting plasma insulin levels, but not with fasting glucose levels. Additionally, specific cross-omic relationships, particularly among proteins and lipids, were accentuated or activated in the case group but not the control group. CONCLUSIONS: Overall, we applied orthogonal, complementary profiling techniques to uncover an inflammatory response linked to elevated triglyceride levels shortly after a GDM pregnancy, which is more pronounced in women who progress to overt diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Diabetes Gestacional , Lactante , Embarazo , Femenino , Humanos , Niño , Estudios de Casos y Controles , Proteómica , Glucosa
4.
Blood Cancer Discov ; 3(1): 16-31, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35019858

RESUMEN

Central nervous system (CNS) dissemination of B-precursor acute lymphoblastic leukemia (B-ALL) has poor prognosis and remains a therapeutic challenge. Here we performed targeted DNA sequencing as well as transcriptional and proteomic profiling of paired leukemia-infiltrating cells in the bone marrow (BM) and CNS of xenografts. Genes governing mRNA translation were upregulated in CNS leukemia, and subclonal genetic profiling confirmed this in both BM-concordant and BM-discordant CNS mutational populations. CNS leukemia cells were exquisitely sensitive to the translation inhibitor omacetaxine mepesuccinate, which reduced xenograft leptomeningeal disease burden. Proteomics demonstrated greater abundance of secreted proteins in CNS-infiltrating cells, including complement component 3 (C3), and drug targeting of C3 influenced CNS disease in xenografts. CNS-infiltrating cells also exhibited selection for stemness traits and metabolic reprogramming. Overall, our study identifies targeting of mRNA translation as a potential therapeutic approach for B-ALL leptomeningeal disease. SIGNIFICANCE: Cancer metastases are often driven by distinct subclones with unique biological properties. Here we show that in B-ALL CNS disease, the leptomeningeal environment selects for cells with unique functional dependencies. Pharmacologic inhibition of mRNA translation signaling treats CNS disease and offers a new therapeutic approach for this condition.This article is highlighted in the In This Issue feature, p. 1.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Neoplasias del Sistema Nervioso Central , Neoplasias Meníngeas , Leucemia-Linfoma Linfoblástico de Células Precursoras , Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/patología , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Humanos , Neoplasias Meníngeas/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Biosíntesis de Proteínas/genética , Proteómica
6.
Cell Rep Med ; 2(12): 100470, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-35028611

RESUMEN

Acute lymphoblastic leukemia (ALL) dissemination to the central nervous system (CNS) is a challenging clinical problem whose underlying mechanisms are poorly understood. Here, we show that primary human ALL samples injected into the femora of immunodeficient mice migrate to the skull and vertebral bone marrow and provoke bone lesions that enable passage into the subarachnoid space. Treatment of leukemia xenografted mice with a biologic antagonist of receptor activator of nuclear factor κB ligand (RANKL) blocks this entry route. In addition to erosion of cranial and vertebral bone, samples from individuals with B-ALL also penetrate the blood-cerebrospinal fluid barrier of recipient mice. Co-administration of C-X-C chemokine receptor 4 (CXCR4) and RANKL antagonists attenuate both identified routes of entry. Our findings suggest that targeted RANKL and CXCR4 pathway inhibitors could attenuate routes of leukemia blast CNS invasion and provide benefit for B-ALL-affected individuals.


Asunto(s)
Sistema Nervioso Central/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Animales , Crisis Blástica/patología , Línea Celular Tumoral , Proteínas de Fusión bcr-abl/metabolismo , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ratones Endogámicos NOD , Modelos Biológicos , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Invasividad Neoplásica , Osteoprotegerina/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/sangre , Leucemia-Linfoma Linfoblástico de Células Precursoras B/líquido cefalorraquídeo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Ligando RANK/antagonistas & inhibidores , Ligando RANK/metabolismo , Receptores CXCR4/metabolismo , Columna Vertebral/patología , Espacio Subaracnoideo/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Sci Transl Med ; 12(561)2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938796

RESUMEN

Although most children survive B cell acute lymphoblastic leukemia (B-ALL), they frequently experience long-term, treatment-related health problems, including osteopenia and osteonecrosis. Because some children present with fractures at ALL diagnosis, we considered the possibility that leukemic B cells contribute directly to bone pathology. To identify potential mechanisms of B-ALL-driven bone destruction, we examined the p53 -/-; Rag2 -/-; Prkdcscid/scid triple mutant (TM) mice and p53 -/-; Prkdcscid/scid double mutant (DM) mouse models of spontaneous B-ALL. In contrast to DM animals, leukemic TM mice displayed brittle bones, and the TM leukemic cells overexpressed Rankl, encoding receptor activator of nuclear factor κB ligand. RANKL is a key regulator of osteoclast differentiation and bone loss. Transfer of TM leukemic cells into immunodeficient recipient mice caused trabecular bone loss. To determine whether human B-ALL can exert similar effects, we evaluated primary human B-ALL blasts isolated at diagnosis for RANKL expression and their impact on bone pathology after their transplantation into NOD.Prkdcscid/scidIl2rgtm1Wjl /SzJ (NSG) recipient mice. Primary B-ALL cells conferred bone destruction evident in increased multinucleated osteoclasts, trabecular bone loss, destruction of the metaphyseal growth plate, and reduction in adipocyte mass in these patient-derived xenografts (PDXs). Treating PDX mice with the RANKL antagonist recombinant osteoprotegerin-Fc (rOPG-Fc) protected the bone from B-ALL-induced destruction even under conditions of heavy tumor burden. Our data demonstrate a critical role of the RANK-RANKL axis in causing B-ALL-mediated bone pathology and provide preclinical support for RANKL-targeted therapy trials to reduce acute and long-term bone destruction in these patients.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Ligando RANK , Animales , Linfocitos B , Humanos , Ratones , Ratones Endogámicos NOD , Osteoclastos
8.
Science ; 368(6495): 1122-1127, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32381589

RESUMEN

Immunological memory specific to previously encountered antigens is a cardinal feature of adaptive lymphoid cells. However, it is unknown whether innate myeloid cells retain memory of prior antigenic stimulation and respond to it more vigorously on subsequent encounters. In this work, we show that murine monocytes and macrophages acquire memory specific to major histocompatibility complex I (MHC-I) antigens, and we identify A-type paired immunoglobulin-like receptors (PIR-As) as the MHC-I receptors necessary for the memory response. We demonstrate that deleting PIR-A in the recipient or blocking PIR-A binding to donor MHC-I molecules blocks memory and attenuates kidney and heart allograft rejection. Thus, innate myeloid cells acquire alloantigen-specific memory that can be targeted to improve transplant outcomes.


Asunto(s)
Rechazo de Injerto/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunidad Innata , Memoria Inmunológica , Macrófagos/inmunología , Monocitos/inmunología , Receptores Inmunológicos/fisiología , Animales , Eliminación de Gen , Rechazo de Injerto/genética , Trasplante de Corazón , Trasplante de Riñón , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Receptores Inmunológicos/genética
9.
Cancer Discov ; 10(4): 568-587, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086311

RESUMEN

Disease recurrence causes significant mortality in B-progenitor acute lymphoblastic leukemia (B-ALL). Genomic analysis of matched diagnosis and relapse samples shows relapse often arising from minor diagnosis subclones. However, why therapy eradicates some subclones while others survive and progress to relapse remains obscure. Elucidation of mechanisms underlying these differing fates requires functional analysis of isolated subclones. Here, large-scale limiting dilution xenografting of diagnosis and relapse samples, combined with targeted sequencing, identified and isolated minor diagnosis subclones that initiate an evolutionary trajectory toward relapse [termed diagnosis Relapse Initiating clones (dRI)]. Compared with other diagnosis subclones, dRIs were drug-tolerant with distinct engraftment and metabolic properties. Transcriptionally, dRIs displayed enrichment for chromatin remodeling, mitochondrial metabolism, proteostasis programs, and an increase in stemness pathways. The isolation and characterization of dRI subclones reveals new avenues for eradicating dRI cells by targeting their distinct metabolic and transcriptional pathways before further evolution renders them fully therapy-resistant. SIGNIFICANCE: Isolation and characterization of subclones from diagnosis samples of patients with B-ALL who relapsed showed that relapse-fated subclones had increased drug tolerance and distinct metabolic and survival transcriptional programs compared with other diagnosis subclones. This study provides strategies to identify and target clinically relevant subclones before further evolution toward relapse.


Asunto(s)
Leucemia Mieloide Aguda/genética , Células Clonales , Femenino , Humanos , Masculino , Recurrencia
10.
Mol Cell Oncol ; 8(1): 1835423, 2020 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-33553597

RESUMEN

Skeletal morbidities continue to cause acute and long-term burdens for B-ALL patients underscoring the need to identify the mechanisms underlying these processes and to develop effective therapies. Our recent findings demonstrated that B-ALL cells isolated at patient diagnosis can cause bone destruction and have identified the receptor activator of nuclear factor κ-B (RANK-RANKL) ligand axis as a critical effector of these effects.

11.
Sci Immunol ; 4(32)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30709843

RESUMEN

Microbiome sequence analyses have suggested that changes in gut bacterial composition are associated with autoimmune disease in humans and animal models. However, little is known of the mechanisms through which the gut microbiota influences autoimmune responses to distant tissues. Here, we evaluated systemic antibody responses against cultured human gut bacterial strains to determine whether observed patterns of anticommensal antibody (ACAb) responses are associated with type 1 diabetes (T1D) in two cohorts of pediatric study participants. In the first cohort, ACAb responses in sera collected from participants within 6 months of T1D diagnosis were compared with age-matched healthy controls and also with patients with recent onset Crohn's disease. ACAb responses against multiple bacterial species discriminated among these three groups. In the second cohort, we asked whether ACAb responses present before diagnosis were associated with later T1D development and with HLA genotype in participants who were discordant for subsequent progression to diabetes. Serum IgG2 antibodies against Roseburia faecis and against a bacterial consortium were associated with future T1D diagnosis in an HLA DR3/DR4 haplotype-dependent manner. These analyses reveal associations between antibody responses to intestinal microbes and HLA-DR genotype and islet autoantibody specificity and with a future diagnosis of T1D. Further, we present a platform to investigate antibacterial antibodies in biological fluids that is applicable to studies of autoimmune diseases and responses to therapeutic interventions.


Asunto(s)
Formación de Anticuerpos/inmunología , Autoinmunidad , Diabetes Mellitus Tipo 1/sangre , Microbioma Gastrointestinal/inmunología , Antígeno HLA-DR3/inmunología , Antígeno HLA-DR4/inmunología , Islotes Pancreáticos/inmunología , Adolescente , Anticuerpos Antibacterianos/inmunología , Autoanticuerpos/inmunología , Niño , Clostridiales/inmunología , Diabetes Mellitus Tipo 1/inmunología , Femenino , Estudios de Seguimiento , Genotipo , Haplotipos , Humanos , Masculino , Pronóstico
12.
Nat Immunol ; 19(7): 685-695, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29925984

RESUMEN

Risk factors for most autoimmune diseases are multifactorial genetic variants modified by environmental risk factors. Type 1 diabetes and celiac disease share high-risk HLA haplotypes, and the prevalence of both diseases has increased in many regions during the past half century. Unknown environmental factors are suspected to have increased the disease penetrance. Celiac disease depends on immune responses to dietary gluten, whereas the environmental risk factors for type 1 diabetes are not yet clear. Here, we consider the shared heritable genetic factors and review evidence of the dietary and microbial exposures, particularly in early life, that might influence the pathogenesis of one or both diseases. A deeper mechanistic understanding of the environmental factors responsible for increased risk of these diseases should provide opportunities to manipulate exposure in children carrying defined risk markers and thus prevent and attenuate disease, as well as to identify new therapeutic strategies for patients.


Asunto(s)
Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Animales , Autoantígenos/inmunología , Enfermedad Celíaca/microbiología , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/microbiología , Dieta , Microbioma Gastrointestinal , Antígenos HLA/genética , Humanos , Lactante , Infecciones/complicaciones , Factores de Riesgo
13.
Adv Exp Med Biol ; 1043: 113-140, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29224093

RESUMEN

The relationships between dietary compounds, derivative metabolites, and host metabolism and immunity are controlled by diverse molecular mechanisms. Essential contributions to these dynamics come from the community of microbes (the microbiome) inhabiting the human digestive tract. The composition and function of the microbiome are shaped by available nutrients, and reciprocally, these organisms produce an as yet poorly defined repertoire of molecules that communicate with the epithelial barrier and the mucosal immune system. We present evidence that diet-derived vitamins and lipids regulate immunity and metabolic function and highlight the diverse mechanisms through which these effects are impacted by sex. We discuss exciting new data emerging from studies using high-throughput sequencing technology, specialized mouse models, and bio-specimens, and clinical data from human subjects that have begun to reveal the complexity of these interactions. Also profiled in this chapter are the striking sex differences in pathways by which dietary nutrients and gut microbes modify metabolism, immunity, and immune- and inflammation-mediated diseases. Although the incidence, severity, and therapeutic responses of many autoimmune diseases differ by sex, the molecular mechanisms of these effects remain poorly understood.


Asunto(s)
Dieta , Microbioma Gastrointestinal , Tracto Gastrointestinal , Inmunidad Mucosa , Absorción Intestinal , Animales , Femenino , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Disparidades en el Estado de Salud , Interacciones Huésped-Patógeno , Humanos , Masculino , Modelos Animales , Estado Nutricional , Valor Nutritivo , Caracteres Sexuales , Factores Sexuales , Transducción de Señal
14.
Sci Immunol ; 2(12)2017 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-28783664

RESUMEN

Mice devoid of T, B, and natural killer (NK) cells distinguish between self and allogeneic nonself despite the absence of an adaptive immune system. When challenged with an allograft, they mount an innate response characterized by accumulation of mature, monocyte-derived dendritic cells (DCs) that produce interleukin-12 and present antigen to T cells. However, the molecular mechanisms by which the innate immune system detects allogeneic nonself to generate these DCs are not known. To address this question, we studied the innate response of Rag2-/- γc-/- mice, which lack T, B, and NK cells, to grafts from allogeneic donors. By positional cloning, we identified that donor polymorphism in the gene encoding signal regulatory protein α (SIRPα) is a key modulator of the recipient's innate allorecognition response. Donors that differed from the recipient in one or both Sirpa alleles elicited an innate alloresponse. The response was mediated by binding of donor SIRPα to recipient CD47 and was modulated by the strength of the SIRPα-CD47 interaction. Therefore, sensing SIRPα polymorphism by CD47 provides a molecular mechanism by which the innate immune system distinguishes between self and allogeneic nonself independently of T, B, and NK cells.

15.
Nature ; 544(7651): 493-497, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28424516

RESUMEN

Cancer cells elude anti-tumour immunity through multiple mechanisms, including upregulated expression of ligands for inhibitory immune checkpoint receptors. Phagocytosis by macrophages plays a critical role in cancer control. Therapeutic blockade of signal regulatory protein (SIRP)-α, an inhibitory receptor on macrophages, or of its ligand CD47 expressed on tumour cells, improves tumour cell elimination in vitro and in vivo, suggesting that blockade of the SIRPα-CD47 checkpoint could be useful in treating human cancer. However, the pro-phagocytic receptor(s) responsible for tumour cell phagocytosis is(are) largely unknown. Here we find that macrophages are much more efficient at phagocytosis of haematopoietic tumour cells, compared with non-haematopoietic tumour cells, in response to SIRPα-CD47 blockade. Using a mouse lacking the signalling lymphocytic activation molecule (SLAM) family of homotypic haematopoietic cell-specific receptors, we determined that phagocytosis of haematopoietic tumour cells during SIRPα-CD47 blockade was strictly dependent on SLAM family receptors in vitro and in vivo. In both mouse and human cells, this function required a single SLAM family member, SLAMF7 (also known as CRACC, CS1, CD319), expressed on macrophages and tumour cell targets. In contrast to most SLAM receptor functions, SLAMF7-mediated phagocytosis was independent of signalling lymphocyte activation molecule-associated protein (SAP) adaptors. Instead, it depended on the ability of SLAMF7 to interact with integrin Mac-1 (refs 18, 19, 20) and utilize signals involving immunoreceptor tyrosine-based activation motifs. These findings elucidate the mechanism by which macrophages engulf and destroy haematopoietic tumour cells. They also reveal a novel SAP adaptor-independent function for a SLAM receptor. Lastly, they suggest that patients with tumours expressing SLAMF7 are more likely to respond to SIRPα-CD47 blockade therapy.


Asunto(s)
Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Antígeno de Macrófago-1/metabolismo , Macrófagos/inmunología , Fagocitosis/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Actinas/metabolismo , Animales , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Antígeno CD47/inmunología , Antígeno CD47/metabolismo , Femenino , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/deficiencia
16.
J Immunol ; 198(2): 590-595, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28069754

RESUMEN

Type 1 diabetes (T1D) is characterized by the autoimmune destruction of pancreatic ß cells. The rapid rise in T1D incidence during the past 50 y suggests environmental factors contribute to the disease. The trillion symbiotic microorganisms inhabiting the mammalian gastrointestinal tract (i.e., the microbiota) influence numerous aspects of host physiology. In this study we review the evidence linking perturbations of the gut microbiome to pancreatic autoimmunity. We discuss data from rodent models demonstrating the essential role of the gut microbiota on the development and function of the host's mucosal and systemic immune systems. Furthermore, we review findings from human longitudinal cohort studies examining the influence of environmental and lifestyle factors on microbiota composition and pancreatic autoimmunity. Taken together, these data underscore the requirement for mechanistic studies to identify bacterial components and metabolites interacting with the innate and adaptive immune system, which would set the basis for preventative or therapeutic strategies in T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Microbioma Gastrointestinal/inmunología , Animales , Humanos
17.
Pediatr Diabetes ; 17(7): 469-477, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27484959

RESUMEN

The prevalence of type 1 and type 2 diabetes have both risen dramatically over the last 50 years. Recent findings point towards the gut microbiota as a potential contributor to these trends. The hundred trillion bacteria residing in the mammalian gut have established a symbiotic relation with their host and influence many aspects of host metabolism, physiology, and immunity. In this review, we examine recent data linking gut microbiome composition and function to anti-pancreatic immunity, insulin-resistance, and obesity. Studies in rodents and human longitudinal studies suggest that an altered gut microbiome characterized by lower diversity and resilience is associated with type 1 and type 2 diabetes. Through its metabolites and enzymatic arsenal, the microbiota shape host metabolism, energy extracted from the diet and contribute to the normal development of the immune system and to tissue inflammation. Increasing evidence underscores the importance of the maternal microbiome, the gestational environment and the conditions of newborn delivery in establishing the gut microbiota of the offspring. Perturbations of the maternal microbiome during gestation, or that of the offspring during early infant development may promote a pro-inflammatory environment conducive to the development of autoimmunity and metabolic disturbance. Collectively the findings reviewed herein underscore the need for mechanistic investigations in rodent models and in human studies to better define the relationships between microbial and host inflammatory activity in diabetes, and to evaluate the potential of microbe-derived therapeutics in the prevention and treatment of both forms of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/microbiología , Diabetes Mellitus Tipo 2/microbiología , Susceptibilidad a Enfermedades/microbiología , Microbioma Gastrointestinal/fisiología , Autoinmunidad/fisiología , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 2/epidemiología , Humanos , Inflamación/inmunología , Inflamación/microbiología , Obesidad/complicaciones , Obesidad/inmunología , Obesidad/microbiología , Factores de Riesgo
18.
J Autoimmun ; 71: 10-8, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26908163

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease resulting from T cell-mediated destruction of the insulin-secreting pancreatic beta cells. During the past 50 years T1D incidence has increased dramatically in many countries accompanied by an earlier age of onset especially in persons with lower genetic risk. These observations have prompted investigations of dynamic environmental factors that may contributor to risk for anti-pancreatic immunity. The gut and pancreas are anatomically and biochemically linked through the enteroinsular axis, a system in which gut-derived immune and metabolic signals have the potential to evoke effects in the pancreas. The gut microbiome (i.e. the 100 trillion symbiotic microorganisms which inhabit the mammalian gastrointestinal tract) influences numerous aspects of host metabolism, development and immunity. Here we examine recent evidence linking gut microbiome composition and function to pancreatic autoimmunity. Studies in children with genetic risk factors for T1D and analyses of the microbiome in rodent models have begun to associations between an altered microbiome composition potentially favoring a pro-inflammatory intestinal metabolic milieu and T1D. We discuss how environmental factors during critical developmental windows - gestation, birth, weaning and puberty may contribute to T1D risk. For example mode of delivery (vaginal or C-section) and exposure to antibiotics (pre- or post-natally) are two factors that modulate the maternal and/or offspring microbiome and can impact T1D development. Taken together, these emerging data underscore the requirement for longitudinal studies and mechanistic investigations in human subjects and rodent models to identify the basis for microbiome modulation of T1D and to identify biomarkers and therapeutics to improve the delayed onset and prevention of the disease.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Microbioma Gastrointestinal/inmunología , Inmunidad , Animales , Autoinmunidad , Diabetes Mellitus Tipo 1/epidemiología , Disbiosis , Ambiente , Exposición a Riesgos Ambientales , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología
19.
Curr Opin Immunol ; 37: 34-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26460968

RESUMEN

The trillions of microorganisms populating the mammalian mucosal surfaces (i.e. the microbiome) participate in the development and function of the host immune system that acts to balance clearance of pathogens with tolerance of beneficial commensals. Recent advances in mucosal immunology and culture-independent sequencing of microbial communities provide support for the hypothesis that the alterations in commensal microbiota alter the host immune response and can enhance risk for autoimmune disease in distant organs. Further explorations of the host-microbiota relationship will improve our understanding of autoimmune disorders and facilitate the discovery of a bacterial-based immunomodulators.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad , Tolerancia Inmunológica , Microbiota/inmunología , Animales , Enfermedades Autoinmunes/microbiología , Secuenciación de Nucleótidos de Alto Rendimiento , Homeostasis , Interacciones Huésped-Patógeno , Humanos , Inmunidad Mucosa , Inmunomodulación , Riesgo
20.
J Immunol ; 194(12): 5663-73, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25964492

RESUMEN

Type 1 diabetes in the NOD mouse model has been linked to >30 insulin-dependent diabetes (Idd) susceptibility loci. Idd4 on chromosome 11 consists of two subloci, Idd4.1 and Idd4.2. Using congenic analysis of alleles in NOD and NOD-resistant (NOR) mice, we previously defined Idd4.1 as an interval containing >50 genes that controlled expression of genes in the type 1 IFN pathway. In this study, we report refined mapping of Idd4.1 to a 1.1-Mb chromosomal region and provide genomic sequence analysis and mechanistic evidence supporting its role in innate immune regulation of islet-directed autoimmunity. Genetic variation at Idd4.1 was mediated by radiation-sensitive hematopoietic cells, and type 1 diabetes protection conferred by the NOR allele was abrogated in mice treated with exogenous type 1 IFN-ß. Next generation sequence analysis of the full Idd4.1 genomic interval in NOD and NOR strains supported Nlrp1b as a strong candidate gene for Idd4.1. Nlrp1b belongs to the Nod-like receptor (NLR) gene family and contributes to inflammasome assembly, caspase-1 recruitment, and release of IL-1ß. The Nlrp1b of NOR was expressed as an alternative spliced isoform that skips exon 9, resulting in a premature stop codon predicted to encode a truncated protein. Functional analysis of the truncated NOR Nlrp1b protein demonstrated that it was unable to recruit caspase-1 and process IL-1ß. Our data suggest that Idd4.1-dependent protection from islet autoimmunity is mediated by differences in type 1 IFN- and IL-1ß-dependent immune responses resulting from genetic variation in Nlrp1b.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Inflamasomas/genética , Sitios de Carácter Cuantitativo , Alelos , Empalme Alternativo , Animales , Proteínas Reguladoras de la Apoptosis/química , Secuencia de Bases , Mapeo Cromosómico , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Femenino , Estudios de Asociación Genética , Inflamasomas/inmunología , Interferón beta/metabolismo , Interferón beta/farmacología , Masculino , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...