Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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.

10.
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
11.
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
12.
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
13.
Acta Ophthalmol ; 93(3): e218-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25271003

RESUMEN

PURPOSE: To verify the effect of vasostatin-1 (VS-1), an anti-angiogenic fragment of chromogranin A, in the prevention of choroidal neovascularization (CNV) in an established mouse model of laser-induced ocular neovascularization. METHODS: Bruch's membrane, the innermost layer of the choroid, was broken by laser photocoagulation in C57/Bl6 mice, to induce CNV. Mice were then treated daily for 14 days by intraperitoneal injection of VS-1 or vehicle (6 mice/group). CNV and vascular leakage were measured at three time-points (day 0, 7 and 14) in vivo by spectral domain optical coherence tomography (OCT) and fluorescein angiography (FA). Ex vivo analysis of CNV was also performed at day 14 by confocal microscopy analysis of dextran-perfused choroidal flat-mounts. RESULTS: In vivo analyses showed that VS-1 significantly reduced CNV at day 14 (p = 0.03) and vascular leakage at day 7 (p = 0.01) and 14 (p = 0.04). Ex vivo confocal microscopy analysis of CNV performed on dextran-perfused choroidal flat-mounts at day 14 confirmed the protective activity of VS-1 (p = 0.01). A significant correlation between the results of in vivo and ex vivo analyses of CNV was also observed (p = 0.001, R(2) = 0.81). CONCLUSION: The results indicate that VS-1 can prevent CNV and vascular leakage in a mouse model of ocular neovascularization, suggesting that this polypeptide might have therapeutic activity in human ocular diseases that are complicated by neovascularization or excessive vascular permeability.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neovascularización Coroidal/tratamiento farmacológico , Cromogranina A/farmacología , Modelos Animales de Enfermedad , Fragmentos de Péptidos/farmacología , Animales , Lámina Basal de la Coroides/cirugía , Permeabilidad Capilar , Neovascularización Coroidal/diagnóstico , Neovascularización Coroidal/etiología , Angiografía con Fluoresceína , Inyecciones Intraperitoneales , Coagulación con Láser/efectos adversos , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Tomografía de Coherencia Óptica
14.
Pharmacol Res ; 98: 69-75, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25014184

RESUMEN

Islet transplantation has been demonstrated to improve glycometabolic control, to reduce hypoglycemic episodes and to halt the progression of diabetic complications. However, the exhaustion of islet function and the side effects related to chronic immunosuppression limit the spread of this technique. Consequently, new immunoregulatory protocols have been developed, with the aim to avoid the use of a life-time immunosuppression. Several approaches have been tested in preclinical models, and some are now under clinical evaluation. The development of new small molecules and new monoclonal or polyclonal antibodies is continuous and raises the possibility of targeting new costimulatory pathways or depleting particular cell types. The use of stem cells and regulatory T cells is underway to take advantage of their immunological properties and to induce tolerance. Xenograft islet transplantation, although having severe problems in terms of immunological compatibility, could theoretically provide an unlimited source of donors; using pigs carrying human immune antigens has showed indeed promising results. A completely different approach, the use of encapsulated islets, has been developed; synthetic structures are used to hide islet alloantigen from the immune system, thus preserving islet endocrine function. Once one of these strategies is demonstrated safe and effective, it will be possible to establish clinical islet transplantation as a treatment for patients with type 1 diabetes long before the onset of diabetic-related complications.


Asunto(s)
Terapia de Inmunosupresión/métodos , Inmunosupresores/uso terapéutico , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Animales , Diabetes Mellitus Tipo 1/terapia , Xenoinjertos , Humanos , Porcinos , Trasplante Autólogo
15.
Retina ; 34(9): 1802-10, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24736462

RESUMEN

BACKGROUND: Choroidal neovascularization (CNV) is the main cause of vision loss in age-related macular degeneration (AMD). In experimental CNV, endothelial progenitor cells (EPCs) contribute to the formation of new vessels. The aim of this study was to investigate whether the behavior of EPCs in patients with AMD supports a role for EPCs in human CNV. METHODS: The number of circulating EPCs that are considered pure endothelial precursors and EPCs with monocytic characteristics, and the plasma levels of regulatory cytokines were evaluated in 23 patients with AMD with active CNV and 20 matched controls. In the patients, this profile was re-evaluated after ranibizumab. RESULTS: When compared with controls, the patients with AMD showed a lower number of both EPC types (P = 0.03) and higher plasma levels (P = 0.03) of stromal cell-derived factor 1. Three monthly injections of ranibizumab returned to control levels the number of circulating EPCs considered pure endothelial precursors and of stromal cell-derived factor 1, but not of monocytic EPCs. CONCLUSION: The observations indicate responsiveness of circulating EPCs to the CNV process in AMD. They suggest the hypothesis that increased stromal cell-derived factor 1 production at the CNV site (reflected in higher plasma levels) recruits EPCs from the circulation, and that antivascular endothelial growth factor therapy selectively decreases the recruitment of cells to be incorporated into new vessels.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Células Progenitoras Endoteliales/fisiología , Degeneración Macular/tratamiento farmacológico , Anciano , Antígenos CD34/sangre , Quimiocina CXCL12/sangre , Neovascularización Coroidal/fisiopatología , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Inyecciones Intravítreas , Antígenos Comunes de Leucocito/sangre , Receptores de Lipopolisacáridos/sangre , Degeneración Macular/fisiopatología , Masculino , Ranibizumab , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre
16.
Exp Diabetes Res ; 2012: 728325, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22566995

RESUMEN

Although cellular and molecular bases of proliferative diabetic retinopathy are only partially understood, it is evident that this complication of diabetes is characterized by the formation of new vessels inside the retina showing abnormal architecture and permeability. This process, if not controlled by selective laser photocoagulation, leads to irreversible retinal damages and loss of vision. Angiogenesis, that is, the condition characterized by the growth of new blood vessels originated from preexisting ones, was shown to have a major role in the pathogenesis of proliferative retinopathy and, as a consequence, intravitreal antiangiogenic injection was suggested as a feasible treatment for this disease. Here, we describe the different antiangiogenic approaches used to treat this disease along with the respective advantages and limitations when compared to laser treatment. Altogether, even though further and longer studies are still needed to clarify the best possible therapeutic protocol, the antiangiogenic treatment will reasonably have a future role in the therapy and prevention of proliferative diabetic retinopathy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Retinopatía Diabética/tratamiento farmacológico , Neovascularización Retiniana/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Retinopatía Diabética/etiología , Humanos
18.
J Am Soc Nephrol ; 17(4 Suppl 2): S123-6, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16565235

RESUMEN

In the past few years, a number of studies have shown that stem cells can be found in virtually every organ of the adult organism. The kidney is not an exception, and resident stem cells have been identified both in the papilla and along the tubules. Of interest, kidney-bound stem cells have been identified also in the bone marrow. When injected, both resident and bone marrow-derived stem cells are able to reach the injured renal tissue and, once there, to differentiate into renal cells. The evidence that, in humans, some of the acute and most of the chronic renal damages lead to ESRD suggests that in normal conditions, the reservoir of stem cells (considering both resident and bone marrow-derived stem cells) is insufficient to allow a major renal regeneration. Probably the number of stem cells that are ready to intervene in an adult kidney are sufficient to compensate for the normal cell turnover but largely inadequate to counteract a major injury. This is confirmed further by the finding that, even by transplanting a syngenic bone marrow in rats with ablation of 5/6 of the renal function, it is not possible to increase the life expectancy of the animals. Altogether, this evidence suggests that, to clarify the potentiality of a stem cell therapy for renal diseases, experiments that aim to clarify the ideal concentration of stem cells to be injected and to identify the best way of administration are needed.


Asunto(s)
Fallo Renal Crónico/terapia , Riñón/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Trasplante de Médula Ósea , Creatinina/sangre , Modelos Animales de Enfermedad , Humanos , Fallo Renal Crónico/metabolismo , Ratas , Regeneración , Factores de Tiempo
19.
Immunogenetics ; 58(2-3): 107-12, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16508738

RESUMEN

Vascular endothelial growth factor (VEGF) is a multifunctional cytokine originally described as an angiogenic factor. A number of reports have recently demonstrated that VEGF increases pancreatic islet survival after islet transplantation by stimulating angiogenesis and improving islet revascularization. Whether VEGF can protect from the autoimmune destruction of insulin-producing beta-cells that characterizes the development of type 1 diabetes is presently unknown. To clarify this issue, we studied the association of three polymorphisms of the promoter region of VEGF with type 1 diabetes in the Italian and the Finnish populations. The polymorphisms considered [C(-2578)A, G(-1190)A, and G(-1154)A] are known to modulate in vitro and in vivo VEGF expression. We found that VEGF promoter genotypes are associated with type 1 diabetes in both populations, but with different combinations. In Italian individuals, the -2578AA and -1190AA genotypes are associated with type 1 diabetes and accelerate its onset, while in Finnish individuals, -1154GG and -1190GG protect from type 1 diabetes and delay its onset. In conclusion, because the expected functional consequence of both genotype combinations is a reduced VEGF expression in diabetic patients, we propose a protective role of VEGF in the development of type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Factor A de Crecimiento Endotelial Vascular/genética , Adulto , Edad de Inicio , Anciano , Regulación hacia Abajo , Femenino , Finlandia , Genotipo , Humanos , Italia , Masculino , Persona de Mediana Edad , Polimorfismo Genético , Regiones Promotoras Genéticas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA