Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Cell Metab ; 36(6): 1302-1319.e12, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38838642

RESUMEN

Glucagon-like peptide-1 receptor (GLP-1R) is a key regulator of glucose metabolism known to be expressed by pancreatic ß cells. We herein investigated the role of GLP-1R on T lymphocytes during immune response. Our data showed that a subset of T lymphocytes expresses GLP-1R, which is upregulated during alloimmune response, similarly to PD-1. When mice received islet or cardiac allotransplantation, an expansion of GLP-1Rpos T cells occurred in the spleen and was found to infiltrate the graft. Additional single-cell RNA sequencing (scRNA-seq) analysis conducted on GLP-1Rpos and GLP-1Rneg CD3+ T cells unveiled the existence of molecular and functional dissimilarities between both subpopulations, as the GLP-1Rpos are mainly composed of exhausted CD8 T cells. GLP-1R acts as a T cell-negative costimulatory molecule, and GLP-1R signaling prolongs allograft survival, mitigates alloimmune response, and reduces T lymphocyte graft infiltration. Notably, GLP-1R antagonism triggered anti-tumor immunity when tested in a preclinical mouse model of colorectal cancer.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón , Trasplante de Islotes Pancreáticos , Ratones Endogámicos C57BL , Animales , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Masculino , Trasplante de Corazón , Ratones Endogámicos BALB C , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Supervivencia de Injerto/inmunología
2.
Front Endocrinol (Lausanne) ; 15: 1306127, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38318298

RESUMEN

Pancreatic beta cells replenishment is considered the next therapeutic option for type 1 diabetes; while stimulating endogenous beta cells proliferation is the "holy grail" for those patients with exhausted beta cell mass. Here we are demonstrating that the pro-apoptotic receptor TMEM219 is expressed in fetal pancreas, in beta cell precursors and in in vitro embryonic-derived endocrine progenitors. TMEM219 signaling negatively regulates beta cells at early stages and induces Caspase 8-mediated cell death. Pharmacological blockade of TMEM219 further rescued beta cell precursor and proliferation markers, and decreased cell death, both in islets and in in vitro-derived endocrine progenitors, allowing for beta cell preservation. While addressing the upstream controlling TMEM219 expression, we determined the TMEM219 miRNet; indeed, one of those miRNAs, miR-129-2, is highly expressed in human islets, particularly in patients at risk or with established type 1 diabetes. miR-129-2 mimic downregulated TMEM219 expression in islets, in in vitro embryonic-derived endocrine progenitors and in highly proliferating insulinoma-derived cells. Moreover, miR-129-2 inhibitor induced a TMEM219 overexpression in insulinoma-derived cells, which restored cell proliferation and functional markers, thus acting as endogenous regulator of TMEM219 expression. The TMEM219 upstream regulator miR129-2 controls the fate of beta cell precursors and may unleash their regenerative potentials to replenish beta cells in type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Insulinoma , MicroARNs , Neoplasias Pancreáticas , Humanos , Proliferación Celular , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Pancreáticas/metabolismo , Factores de Transcripción/metabolismo
3.
Pharmacol Res ; 190: 106710, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36871895

RESUMEN

Diabetic kidney disease (DKD) is the first cause of end-stage kidney disease in patients with diabetes and its prevalence is increasing worldwide. It encompasses histological alterations that mainly affect the glomerular filtration unit, which include thickening of the basement membrane, mesangial cell proliferation, endothelial alteration, and podocyte injury. These morphological abnormalities further result in a persistent increase of urinary albumin-to-creatinine ratio and in a reduction of the estimated glomerular filtration rate. Several molecular and cellular mechanisms have been recognized, up to date, as major players in mediating such clinical and histological features and many more are being under investigation. This review summarizes the most recent advances in understanding cell death mechanisms, intracellular signaling pathways and molecular effectors that play a role in the onset and progression of diabetic kidney damage. Some of those molecular and cellular mechanisms have been already successfully targeted in preclinical models of DKD and, in some cases, strategies have been tested in clinical trials. Finally, this report sheds light on the relevance of novel pathways that may become therapeutic targets for future applications in DKD.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Podocitos , Humanos , Nefropatías Diabéticas/metabolismo , Podocitos/patología , Transducción de Señal , Tasa de Filtración Glomerular , Diabetes Mellitus/metabolismo
4.
Acta Diabetol ; 59(9): 1157-1167, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35729357

RESUMEN

AIMS: Abnormalities in the oculomotor system may represent an early sign of diabetic neuropathy and are currently poorly studied. We designed an eye-tracking-based test to evaluate oculomotor function in patients with type 1 diabetes. METHODS: We used the SRLab-Tobii TX300 Eye tracker®, an eye-tracking device, coupled with software that we developed to test abnormalities in the oculomotor system. The software consists of a series of eye-tracking tasks divided into 4 classes of parameters (Resistance, Wideness, Pursuit and Velocity) to evaluate both smooth and saccadic movement in different directions. We analyzed the oculomotor system in 34 healthy volunteers and in 34 patients with long-standing type 1 diabetes. RESULTS: Among the 474 parameters analyzed with the eye-tracking-based system, 11% were significantly altered in patients with type 1 diabetes (p < 0.05), with a higher proportion of abnormalities observed in the Wideness (24%) and Resistance (10%) parameters. Patients with type 1 diabetes without diabetic neuropathy showed more frequently anomalous measurements in the Resistance class (p = 0.02). The classes of Velocity and Pursuit were less frequently altered in patients with type 1 diabetes as compared to healthy subjects, with anomalous measurements mainly observed in patients with diabetic neuropathy. CONCLUSIONS: Abnormalities in oculomotor system function can be detected in patients with type 1 diabetes using a novel eye-tracking-based test. A larger cohort study may further determine thresholds of normality and validate whether eye-tracking can be used to non-invasively characterize early signs of diabetic neuropathy. TRIAL: NCT04608890.


Asunto(s)
Diabetes Mellitus Tipo 1 , Neuropatías Diabéticas , Estudios de Cohortes , Diabetes Mellitus Tipo 1/complicaciones , Neuropatías Diabéticas/diagnóstico , Neuropatías Diabéticas/etiología , Humanos , Seguimiento Ocular Uniforme , Movimientos Sacádicos
5.
Diabetes ; 71(7): 1579-1590, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35499468

RESUMEN

Recent studies have shown that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may induce metabolic distress, leading to hyperglycemia in patients affected by coronavirus disease 19 (COVID-19). We investigated the potential indirect and direct effects of SARS-CoV-2 on human pancreatic islets in 10 patients who became hyperglycemic after COVID-19. Although there was no evidence of peripheral anti-islet autoimmunity, the serum of these patients displayed toxicity on human pancreatic islets, which could be abrogated by the use of anti-interleukin-1ß (IL-1ß), anti-IL-6, and anti-tumor necrosis factor α, cytokines known to be highly upregulated during COVID-19. Interestingly, the receptors of those aforementioned cytokines were highly expressed on human pancreatic islets. An increase in peripheral unmethylated INS DNA, a marker of cell death, was evident in several patients with COVID-19. Pathology of the pancreas from deceased hyperglycemic patients who had COVID-19 revealed mild lymphocytic infiltration of pancreatic islets and pancreatic lymph nodes. Moreover, SARS-CoV-2-specific viral RNA, along with the presence of several immature insulin granules or proinsulin, was detected in postmortem pancreatic tissues, suggestive of ß-cell-altered proinsulin processing, as well as ß-cell degeneration and hyperstimulation. These data demonstrate that SARS-CoV-2 may negatively affect human pancreatic islet function and survival by creating inflammatory conditions, possibly with a direct tropism, which may in turn lead to metabolic abnormalities observed in patients with COVID-19.


Asunto(s)
COVID-19 , Islotes Pancreáticos , COVID-19/complicaciones , Citocinas/metabolismo , Humanos , Hiperglucemia/virología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/virología , Proinsulina/metabolismo , SARS-CoV-2
6.
Diabetes ; 71(8): 1800-1806, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35551366

RESUMEN

Patients with type 1 diabetes (T1D) may develop severe outcomes during coronavirus disease 2019 (COVID-19), but their ability to generate an immune response against the SARS-CoV-2 mRNA vaccines remains to be established. We evaluated the safety, immunogenicity, and glycometabolic effects of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccines in patients with T1D. A total of 375 patients (326 with T1D and 49 subjects without diabetes) who received two doses of the SARS-CoV-2 mRNA vaccines (mRNA-1273, BNT162b2) between March and April 2021 at ASST Fatebenefratelli Sacco were included in this monocentric observational study. Local and systemic adverse events were reported in both groups after SARS-CoV-2 mRNA vaccination, without statistical differences between them. While both patients with T1D and subjects without diabetes exhibited a parallel increase in anti-SARS-CoV-2 spike titers after vaccination, the majority of patients with T1D (70% and 78%, respectively) did not show any increase in the SARS-CoV-2-specific cytotoxic response compared with the robust increase observed in all subjects without diabetes. A reduced secretion of the T-cell-related cytokines interleukin-2 and tumor necrosis factor-α in vaccinated patients with T1D was also observed. No glycometabolic alterations were evident in patients with T1D using continuous glucose monitoring during follow-up. Administration of the SARS-CoV-2 mRNA vaccine is associated with an impaired cellular SARS-CoV-2-specific cytotoxic immune response in patients with T1D.


Asunto(s)
Vacuna nCoV-2019 mRNA-1273 , Vacuna BNT162 , Diabetes Mellitus Tipo 1 , Vacuna nCoV-2019 mRNA-1273/efectos adversos , Vacuna nCoV-2019 mRNA-1273/inmunología , Anticuerpos Antivirales , Vacuna BNT162/efectos adversos , Vacuna BNT162/inmunología , Glucemia , Automonitorización de la Glucosa Sanguínea , COVID-19/prevención & control , Estudios de Cohortes , Diabetes Mellitus Tipo 1/inmunología , Humanos
7.
Nat Commun ; 13(1): 684, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115561

RESUMEN

Loss of pancreatic beta cells is a central feature of type 1 (T1D) and type 2 (T2D) diabetes, but a therapeutic strategy to preserve beta cell mass remains to be established. Here we show that the death receptor TMEM219 is expressed on pancreatic beta cells and that signaling through its ligand insulin-like growth factor binding protein 3 (IGFBP3) leads to beta cell loss and dysfunction. Increased peripheral IGFBP3 was observed in established and at-risk T1D/T2D patients and was confirmed in T1D/T2D preclinical models, suggesting that dysfunctional IGFBP3/TMEM219 signaling is associated with abnormalities in beta cells homeostasis. In vitro and in vivo short-term IGFBP3/TMEM219 inhibition and TMEM219 genetic ablation preserved beta cells and prevented/delayed diabetes onset, while long-term IGFBP3/TMEM219 blockade allowed for beta cell expansion. Interestingly, in several patients' cohorts restoration of appropriate IGFBP3 levels was associated with improved beta cell function. The IGFBP3/TMEM219 pathway is thus shown to be a physiological regulator of beta cell homeostasis and is also demonstrated to be disrupted in T1D/T2D. IGFBP3/TMEM219 targeting may therefore serve as a therapeutic option in diabetes.


Asunto(s)
Regulación de la Expresión Génica , Homeostasis/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Células Secretoras de Insulina/metabolismo , Proteínas de la Membrana/genética , Transducción de Señal/genética , Adulto , Animales , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Humanos , Immunoblotting , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
JCI Insight ; 6(24)2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34784300

RESUMEN

A substantial proportion of patients who have recovered from coronavirus disease-2019 (COVID-19) experience COVID-19-related symptoms even months after hospital discharge. We extensively immunologically characterized patients who recovered from COVID-19. In these patients, T cells were exhausted, with increased PD-1+ T cells, as compared with healthy controls. Plasma levels of IL-1ß, IL-1RA, and IL-8, among others, were also increased in patients who recovered from COVID-19. This altered immunophenotype was mirrored by a reduced ex vivo T cell response to both nonspecific and specific stimulation, revealing a dysfunctional status of T cells, including a poor response to SARS-CoV-2 antigens. Altered levels of plasma soluble PD-L1, as well as of PD1 promoter methylation and PD1-targeting miR-15-5p, in CD8+ T cells were also observed, suggesting abnormal function of the PD-1/PD-L1 immune checkpoint axis. Notably, ex vivo blockade of PD-1 nearly normalized the aforementioned immunophenotype and restored T cell function, reverting the observed post-COVID-19 immune abnormalities; indeed, we also noted an increased T cell-mediated response to SARS-CoV-2 peptides. Finally, in a neutralization assay, PD-1 blockade did not alter the ability of T cells to neutralize SARS-CoV-2 spike pseudotyped lentivirus infection. Immune checkpoint blockade ameliorates post-COVID-19 immune abnormalities and stimulates an anti-SARS-CoV-2 immune response.


Asunto(s)
COVID-19/complicaciones , Citocinas/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Receptor de Muerte Celular Programada 1/inmunología , SARS-CoV-2/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Antígeno B7-H1/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Estudios de Casos y Controles , Citocinas/efectos de los fármacos , Metilación de ADN , Femenino , Humanos , Inmunofenotipificación , Técnicas In Vitro , Proteína Antagonista del Receptor de Interleucina 1/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/inmunología , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/inmunología , Interleucina-8/efectos de los fármacos , Interleucina-8/inmunología , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Regiones Promotoras Genéticas , Síndrome Post Agudo de COVID-19
9.
Front Immunol ; 12: 694118, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34305929

RESUMEN

Despite the increasing knowledge of pathophysiological mechanisms underlying the onset of type 1 diabetes (T1D), the quest for therapeutic options capable of delaying/reverting the diseases is still ongoing. Among all strategies currently tested in T1D, the use of hematopoietic stem cell (HSC)-based approaches and of teplizumab, showed the most encouraging results. Few clinical trials have already demonstrated the beneficial effects of HSCs in T1D, while the durability of the effect is yet to be established. Investigators are also trying to understand whether the use of selected and better-characterized HSCs subsets may provide more benefits with less risks. Interestingly, ex vivo manipulated HSCs showed promising results in murine models and the recent introduction of the humanized mouse models accelerated the translational potentials of such studies and their final road to clinic. Indeed, immunomodulatory as well as trafficking abilities can be enhanced in genetically modulated HSCs and genetically engineered HSCs may be viewed as a novel "biologic" therapy, to be further tested and explored in T1D and in other autoimmune/immune-related disorders.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Diabetes Mellitus Tipo 1/terapia , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Hipoglucemiantes/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados/efectos adversos , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Hipoglucemiantes/efectos adversos , Ratones , Fenotipo , Resultado del Tratamiento
10.
Metabolism ; 121: 154804, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34097917

RESUMEN

AIMS/HYPOTHESIS: Inflammation has a major role in diabetic kidney disease. We thus investigated the role of the IL-8-CXCR1/2 axis in favoring kidney damage in diabetes. METHODS: Urinary IL-8 levels were measured in 1247 patients of the Joslin Kidney Study in type 2 diabetes (T2D). The expression of IL-8 and of its membrane receptors CXCR1/CXCR2 was quantified in kidney tissues in patients with T2D and in controls. The effect of CXCR1/2 blockade on diabetic kidney disease was evaluated in db/db mice. RESULTS: IL-8 urinary levels were increased in patients with T2D and diabetic kidney disease, with the highest urinary IL-8 levels found in the patients with the largest decline in glomerular filtration rate, with an increased albumin/creatine ratio and the worst renal outcome. Moreover, glomerular IL-8 renal expression was increased in patients with T2D, as compared to controls. High glucose elicits abundant IL-8 secretion in cultured human immortalized podocytes in vitro. Finally, in diabetic db/db mice and in podocytes in vitro, CXCR1/2 blockade mitigated albuminuria, reduced mesangial expansion, decreased podocyte apoptosis and reduced DNA damage. CONCLUSIONS/INTERPRETATION: The IL-8- CXCR1/2 axis may have a role in diabetic kidney disease by inducing podocyte damage. Indeed, targeting the IL-8-CXCR1/2 axis may reduce the burden of diabetic kidney disease.


Asunto(s)
Nefropatías Diabéticas/genética , Interleucina-8/fisiología , Receptores CXCR/fisiología , Adulto , Animales , Estudios de Casos y Controles , Células Cultivadas , Estudios de Cohortes , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Italia , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Podocitos/metabolismo , Podocitos/patología , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Transducción de Señal/fisiología
11.
Nat Metab ; 3(6): 774-785, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34035524

RESUMEN

Patients with coronavirus disease 2019 (COVID-19) are reported to have a greater prevalence of hyperglycaemia. Cytokine release as a consequence of severe acute respiratory syndrome coronavirus 2 infection may precipitate the onset of metabolic alterations by affecting glucose homeostasis. Here we describe abnormalities in glycometabolic control, insulin resistance and beta cell function in patients with COVID-19 without any pre-existing history or diagnosis of diabetes, and document glycaemic abnormalities in recovered patients 2 months after onset of disease. In a cohort of 551 patients hospitalized for COVID-19 in Italy, we found that 46% of patients were hyperglycaemic, whereas 27% were normoglycaemic. Using clinical assays and continuous glucose monitoring in a subset of patients, we detected altered glycometabolic control, with insulin resistance and an abnormal cytokine profile, even in normoglycaemic patients. Glycaemic abnormalities can be detected for at least 2 months in patients who recovered from COVID-19. Our data demonstrate that COVID-19 is associated with aberrant glycometabolic control, which can persist even after recovery, suggesting that further investigation of metabolic abnormalities in the context of long COVID is warranted.


Asunto(s)
Glucemia/metabolismo , COVID-19/sangre , Hiperglucemia/metabolismo , COVID-19/complicaciones , COVID-19/virología , Estudios de Cohortes , Humanos , Hiperglucemia/complicaciones , Resistencia a la Insulina , Células Secretoras de Insulina/patología , SARS-CoV-2/aislamiento & purificación
12.
Am J Transplant ; 21(10): 3280-3295, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33764625

RESUMEN

Despite much progress in improving graft outcome during cardiac transplantation, chronic allograft vasculopathy (CAV) remains an impediment to long-term graft survival. MicroRNAs (miRNAs) emerged as regulators of the immune response. Here, we aimed to examine the miRNA network involved in CAV. miRNA profiling of heart samples obtained from a murine model of CAV and from cardiac-transplanted patients with CAV demonstrated that miR-21 was most significantly expressed and was primarily localized to macrophages. Interestingly, macrophage depletion with clodronate did not significantly prolong allograft survival in mice, while conditional deletion of miR-21 in macrophages or the use of a specific miR-21 antagomir resulted in indefinite cardiac allograft survival and abrogated CAV. The immunophenotype, secretome, ability to phagocytose, migration, and antigen presentation of macrophages were unaffected by miR-21 targeting, while macrophage metabolism was reprogrammed, with a shift toward oxidative phosphorylation in naïve macrophages and with an inhibition of glycolysis in pro-inflammatory macrophages. The aforementioned effects resulted in an increase in M2-like macrophages, which could be reverted by the addition of L-arginine. RNA-seq analysis confirmed alterations in arginase-associated pathways associated with miR-21 antagonism. In conclusion, miR-21 is overexpressed in murine and human CAV, and its targeting delays CAV onset by reprogramming macrophages metabolism.


Asunto(s)
Trasplante de Corazón , MicroARNs , Aloinjertos , Animales , Rechazo de Injerto/genética , Rechazo de Injerto/prevención & control , Trasplante de Corazón/efectos adversos , Humanos , Macrófagos , Ratones , MicroARNs/genética
13.
Artículo en Inglés | MEDLINE | ID: mdl-33188009

RESUMEN

INTRODUCTION: Gestational diabetes mellitus (GDM) is the most frequent metabolic complication during pregnancy and is associated with development of short-term and long-term complications for newborns, with large-for-gestational-age (LGA) being particularly common. Interestingly, the mechanism behind altered fetal growth in GDM is only partially understood. RESEARCH DESIGN AND METHODS: A proteomic approach was used to analyze placental samples obtained from healthy pregnant women (n=5), patients with GDM (n=12) and with GDM and LGA (n=5). Effects of altered proteins on fetal development were tested in vitro in human embryonic stem cells (hESCs). RESULTS: Here, we demonstrate that the placental proteome is altered in pregnant women affected by GDM with LGA, with at least 37 proteins differentially expressed to a higher degree (p<0.05) as compared with those with GDM but without LGA. Among these proteins, 10 are involved in regulating tissue differentiation and/or fetal growth and development, with bone marrow proteoglycan (PRG2) and dipeptidyl peptidase-4 (DPP-4) being highly expressed. Both PRG2 and DPP-4 altered the transcriptome profile of stem cells differentiation markers when tested in vitro in hESCs, suggesting a potential role in the onset of fetal abnormalities. CONCLUSIONS: Our findings suggest that placental dysfunction may be directly responsible for abnormal fetal growth/development during GDM. Once established on a larger population, inhibitors of the pathways involving those altered factors may be tested in conditions such as GDM and LGA, in which therapeutic approaches are still lacking.


Asunto(s)
Diabetes Gestacional , Macrosomía Fetal , Proteoma , Cesárea , Femenino , Macrosomía Fetal/genética , Humanos , Recién Nacido , Embarazo , Proteoma/genética , Proteómica
14.
Eur J Public Health ; 30(1): 70-75, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31263891

RESUMEN

BACKGROUND: The age around 50 years represents a crucial point for women: menopause leads to biological changes and it begins breast and colon-rectal cancer screening. This study aimed at assessing frequencies of cardiovascular risk factors and analyzing participation in screening and vaccination. METHODS: In 2017, a cross-sectional study was performed in Northern Italy. Totally, 12 249 women, aged between 50 and 54 years, were enrolled by General Practitioners (GPs). It was used a 21-item form, with information about: socio-demographic, anamnestic and clinical data, execution of a booster shot of tetanus-diphtheria-acellular pertussis (Tdap) vaccine in the last decade and of PAP-test, mammography and faecal occult blood test in the last 2 years. Descriptive and crosstab χ2 analyses were performed with STATA MP13. The significance level was P ≤ 0.05. RESULTS: Our findings showed the presence of cardiovascular risk factors, such as obesity (10.95%), hypertension (13.76%), hyperlipidaemia (11.57%), glycaemia ≥ 100 mg dl-1 (16.97%), poor physical activity (73.49%), smoking (18.28%), cardiovascular family history (FH) (51.70%). There were a lower participation in colo-rectal cancer screening (45.09%) compared with breast (85.06%) and cervical (77.16%) cancer screening and an insufficient Tdap booster dose compliance (17.56%). Chi-square analyses showed correlations between cardiovascular FH and body mass index, hypertension, hyperlipidaemia, glycaemia and smoking, and between cancer FH and participation in breast and colo-rectal cancer screening (P < 0.05). CONCLUSIONS: Women with cardiovascular disease FH represent a priority target of educational interventions considering the prevalence of concomitant risk factors. Programmes aimed at increasing screening and vaccination participation should be implemented.


Asunto(s)
Anticuerpos Antibacterianos , Vacunas contra Difteria, Tétanos y Tos Ferina Acelular , Enfermedad Crónica , Estudios Transversales , Femenino , Humanos , Italia/epidemiología , Persona de Mediana Edad
15.
Diabetes ; 67(10): 2038-2053, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30065030

RESUMEN

Extracellular ATP (eATP) activates T cells by engaging the P2X7R receptor. We identified two loss-of-function P2X7R mutations that are protective against type 1 diabetes (T1D) and thus hypothesized that eATP/P2X7R signaling may represent an early step in T1D onset. Specifically, we observed that in patients with newly diagnosed T1D, P2X7R is upregulated on CD8+ effector T cells in comparison with healthy control subjects. eATP is released at high levels by human/murine islets in vitro in high-glucose/inflammatory conditions, thus upregulating P2X7R on CD8+ T cells in vitro. P2X7R blockade with oxidized ATP reduces the CD8+ T cell-mediated autoimmune response in vitro and delays diabetes onset in NOD mice. Autoreactive CD8+ T-cell activation is highly dependent upon eATP/P2X7R-mediated priming, while a novel sP2X7R recombinant protein abrogates changes in metabolism and the autoimmune response associated with CD8+ T cells. eATP/P2X7R signaling facilitates the onset of autoimmune T1D by fueling autoreactive CD8+ cells and therefore represents a novel targeted therapeutic for the disorder.


Asunto(s)
Adenosina Trifosfato/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Autoinmunidad/genética , Autoinmunidad/fisiología , Diabetes Mellitus Tipo 1/genética , Femenino , Citometría de Flujo , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Mutación/genética , Receptores Purinérgicos P2X7/genética
16.
J Clin Invest ; 128(8): 3490-3503, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010623

RESUMEN

Purinergic receptor-7 (P2X7R) signaling controls Th17 and Th1 generation/differentiation, while NOD-like receptor P3 (NLRP3) acts as a Th2 transcriptional factor. Here, we demonstrated the existence of a P2X7R/NLRP3 pathway in T cells that is dysregulated by a P2X7R intracellular region loss-of-function mutation, leading to NLRP3 displacement and to excessive Th17 generation due to abrogation of the NLRP3-mediated Th2 program. This ultimately resulted in poor outcomes in cardiac-transplanted patients carrying the mutant allele, who showed abnormal Th17 generation. Transient NLRP3 silencing in nonmutant T cells or overexpression in mutant T cells normalized the Th profile. Interestingly, IL-17 blockade reduced Th17 skewing of human T cells in vitro and abrogated the severe allograft vasculopathy and abnormal Th17 generation observed in preclinical models in which P2X7R was genetically deleted. This P2X7R intracellular region mutation thus impaired the modulatory effects of P2X7R on NLRP3 expression and function in T cells and led to NLRP3 dysregulation and Th17 skewing, delineating a high-risk group of cardiac-transplanted patients who may benefit from personalized therapy.


Asunto(s)
Rechazo de Injerto/inmunología , Trasplante de Corazón , Mutación , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Receptores Purinérgicos P2X7/inmunología , Células Th17/inmunología , Células Th2/inmunología , Aloinjertos , Femenino , Rechazo de Injerto/genética , Rechazo de Injerto/patología , Rechazo de Injerto/terapia , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Medicina de Precisión , Receptores Purinérgicos P2X7/genética , Células Th17/patología , Células Th2/patología
17.
Front Immunol ; 9: 1387, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29971065

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are multipotent stem cells that have been harnessed as a curative therapy for patients with hematological malignancies. Notably, the discovery that HSPCs are endowed with immunoregulatory properties suggests that HSPC-based therapeutic approaches may be used to treat autoimmune diseases. Indeed, infusion with HSPCs has shown promising results in the treatment of type 1 diabetes (T1D) and remains the only "experimental therapy" that has achieved a satisfactory rate of remission (nearly 60%) in T1D. Patients with newly diagnosed T1D have been successfully reverted to normoglycemia by administration of autologous HSPCs in association with a non-myeloablative immunosuppressive regimen. However, this approach is hampered by a high incidence of adverse effects linked to immunosuppression. Herein, we report that while the use of autologous HSPCs is capable of improving C-peptide production in patients with T1D, ex vivo modulation of HSPCs with prostaglandins (PGs) increases their immunoregulatory properties by upregulating expression of the immune checkpoint-signaling molecule PD-L1. Surprisingly, CXCR4 was upregulated as well, which could enhance HSPC trafficking toward the inflamed pancreatic zone. When tested in murine and human in vitro autoimmune assays, PG-modulated HSPCs were shown to abrogate the autoreactive T cell response. The use of PG-modulated HSPCs may thus provide an attractive and novel treatment of autoimmune diabetes.

18.
Sci Transl Med ; 9(416)2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-29141886

RESUMEN

Immunologically based clinical trials performed thus far have failed to cure type 1 diabetes (T1D), in part because these approaches were nonspecific. Because the disease is driven by autoreactive CD4 T cells, which destroy ß cells, transplantation of hematopoietic stem and progenitor cells (HSPCs) has been recently offered as a therapy for T1D. Our transcriptomic profiling of HSPCs revealed that these cells are deficient in programmed death ligand 1 (PD-L1), an important immune checkpoint, in the T1D nonobese diabetic (NOD) mouse model. Notably, the immunoregulatory molecule PD-L1 plays a determinant role in controlling/inhibiting activated T cells and thus maintains immune tolerance. Furthermore, our genome-wide and bioinformatic analysis revealed the existence of a network of microRNAs (miRNAs) controlling PD-L1 expression, and silencing one of key altered miRNAs restored PD-L1 expression in HSPCs. We therefore sought to determine whether restoration of this defect would cure T1D as an alternative to immunosuppression. Genetically engineered or pharmacologically modulated HSPCs overexpressing PD-L1 inhibited the autoimmune response in vitro, reverted diabetes in newly hyperglycemic NOD mice in vivo, and homed to the pancreas of hyperglycemic NOD mice. The PD-L1 expression defect was confirmed in human HSPCs in T1D patients as well, and pharmacologically modulated human HSPCs also inhibited the autoimmune response in vitro. Targeting a specific immune checkpoint defect in HSPCs thus may contribute to establishing a cure for T1D.


Asunto(s)
Antígeno B7-H1/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hiperglucemia/metabolismo , Animales , Autoinmunidad/genética , Autoinmunidad/fisiología , Antígeno B7-H1/genética , Terapia Genética , Humanos , Hiperglucemia/genética , Ratones , Ratones Endogámicos C57BL , Células Madre/citología , Células Madre/metabolismo
19.
Eur J Pharmacol ; 769: 313-23, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26607470

RESUMEN

Although the pathogenesis of diabetic retinopathy (DR) is still insufficiently understood, new evidences indicate 'retinal inflammation' as an important player in the pathogenesis of the complication. Accordingly, common sets of upregulated inflammatory cytokines are found in serum, vitreous and aqueous samples obtained from subjects with DR, and these cytokines can have multiple interactions to impact the pathogenesis of the disease. Thus, based on previously published data, we investigated the effects of Palmitoylethanolamide (PEA), an endogenous lipid amide that belongs to the N-acyl-ethanolamines family, on DR in streptozotocin (STZ)-induced diabetic rats. PEA (10mg/kg) was administered orally daily starting 3 days after the iv administration of STZ. The rats were killed 15 and 60day later and eyes were enucleated to evaluate, through immunohistochemical analysis, the key inflammatory events involved in the breakdown of blood retinal barrier (BRB). Immunohistochemical analysis confirmed the presence of VEGF, ICAM-1, nitrotyrosine (a marker of peroxynitrite), and tight junctions in the retina of STZ-treated rats. Of interest, the extent of injury was significantly reduced after treatment with PEA. Altogether, this study provides the first evidence that PEA attenuates the degree of inflammation while preserving the blood-retinal barrier in rats with experimental DR.


Asunto(s)
Antiinflamatorios/farmacología , Diabetes Mellitus Experimental/complicaciones , Retinopatía Diabética/complicaciones , Retinopatía Diabética/tratamiento farmacológico , Etanolaminas/farmacología , Ácidos Palmíticos/farmacología , Retina/efectos de los fármacos , Amidas , Animales , Antiinflamatorios/uso terapéutico , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Claudinas/metabolismo , Citocinas/metabolismo , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Etanolaminas/uso terapéutico , Inflamación/tratamiento farmacológico , Molécula 1 de Adhesión Intercelular/biosíntesis , Masculino , Ácidos Palmíticos/uso terapéutico , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Retina/metabolismo , Retina/patología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Proteína de la Zonula Occludens-1/metabolismo
20.
Cell Stem Cell ; 17(4): 486-498, 2015 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-26431183

RESUMEN

The role of circulating factors in regulating colonic stem cells (CoSCs) and colonic epithelial homeostasis is unclear. Individuals with long-standing type 1 diabetes (T1D) frequently have intestinal symptoms, termed diabetic enteropathy (DE), though its etiology is unknown. Here, we report that T1D patients with DE exhibit abnormalities in their intestinal mucosa and CoSCs, which fail to generate in vitro mini-guts. Proteomic profiling of T1D+DE patient serum revealed altered levels of insulin-like growth factor 1 (IGF-I) and its binding protein 3 (IGFBP3). IGFBP3 prevented in vitro growth of patient-derived organoids via binding its receptor TMEM219, in an IGF-I-independent manner, and disrupted in vivo CoSC function in a preclinical DE model. Restoration of normoglycemia in patients with long-standing T1D via kidney-pancreas transplantation or in diabetic mice by treatment with an ecto-TMEM219 recombinant protein normalized circulating IGF-I/IGFBP3 levels and reestablished CoSC homeostasis. These findings demonstrate that peripheral IGF-I/IGFBP3 controls CoSCs and their dysfunction in DE.


Asunto(s)
Colon/citología , Complicaciones de la Diabetes/patología , Diabetes Mellitus Experimental/patología , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre/fisiología , Animales , Colon/fisiología , Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus Experimental/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Proteínas de la Membrana/metabolismo , Ratones , Proteómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA