Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 310(11): E1016-26, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27117005

RESUMEN

Long-term pancreatic cold ischemia contributes to decreased islet number and viability after isolation and culture, leading to poor islet transplantation outcome in patients with type 1 diabetes. In this study, we examined mechanisms of pancreatic cold preservation and rewarming-induced injury by interrogating the proapoptotic gene BBC3/Bbc3, also known as Puma (p53 upregulated modulator of apoptosis), using three experimental models: 1) bioluminescence imaging of isolated luciferase-transgenic ("Firefly") Lewis rat islets, 2) cold preservation of en bloc-harvested pancreata from Bbc3-knockout (KO) mice, and 3) cold preservation and rewarming of human pancreata and isolated islets. Cold preservation-mediated islet injury occurred during rewarming in "Firefly" islets. Silencing Bbc3 by transfecting Bbc3 siRNA into islets in vitro prior to cold preservation improved postpreservation mitochondrial viability. Cold preservation resulted in decreased postisolation islet yield in both wild-type and Bbc3 KO pancreata. However, after culture, the islet viability was significantly higher in Bbc3-KO islets, suggesting that different mechanisms are involved in islet damage/loss during isolation and culture. Furthermore, Bbc3-KO islets from cold-preserved pancreata showed reduced HMGB1 (high-mobility group box 1 protein) expression and decreased levels of 4-hydroxynonenal (4-HNE) protein adducts, which was indicative of reduced oxidative stress. During human islet isolation, BBC3 protein was upregulated in digested tissue from cold-preserved pancreata. Hypoxia in cold preservation increased BBC3 mRNA and protein in isolated human islets after rewarming in culture and reduced islet viability. These results demonstrated the involvement of BBC3/Bbc3 in cold preservation/rewarming-mediated islet injury, possibly through modulating HMGB1- and oxidative stress-mediated injury to islets.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Supervivencia Celular/fisiología , Criopreservación/métodos , Islotes Pancreáticos/lesiones , Islotes Pancreáticos/fisiopatología , Proteínas Proto-Oncogénicas/metabolismo , Recalentamiento/efectos adversos , Animales , Células Cultivadas , Humanos , Estrés Oxidativo/fisiología , Ratas , Ratas Endogámicas Lew
2.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G675-G687, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27609771

RESUMEN

Epidemiological studies support strong links between obesity, diabetes, and pancreatic disorders including pancreatitis and pancreatic adenocarcinoma (PDAC). Type 2 diabetes (T2DM) is associated with insulin resistance, hyperglycemia, and hyperinsulinemia, the latter due to increased insulin secretion by pancreatic beta-cells. We reported that high-fat diet-induced PDAC progression in mice is associated with hyperglycemia, hyperinsulinemia, and activation of pancreatic stellate cells (PaSC). We investigated here the effects of high concentrations of insulin and glucose on mouse and human PaSC growth and fibrosing responses. We found that compared with normal, pancreata from T2DM patients displayed extensive collagen deposition and activated PaSC in islet and peri-islet exocrine pancreas. Mice fed a high-fat diet for up to 12 mo similarly displayed increasing peri-islet fibrosis compared with mice fed control diet. Both quiescent and activated PaSC coexpress insulin (IR; mainly A type) and IGF (IGF-1R) receptors, and both insulin and glucose modulate receptor expression. In cultured PaSC, insulin induced rapid tyrosine autophosphorylation of IR/IGF-1R at specific kinase domain activation loop sites, activated Akt/mTOR/p70S6K signaling, and inactivated FoxO1, a transcription factor that restrains cell growth. Insulin did not promote activation of quiescent PaSC in either 5 mM or 25 mM glucose containing media. However, in activated PaSC, insulin enhanced cell proliferation and augmented production of extracellular matrix proteins, and these effects were abolished by specific inhibition of mTORC1 and mTORC2. In conclusion, our data support the concept that increased local glucose and insulin concentrations associated with obesity and T2DM promote PaSC growth and fibrosing responses.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/patología , Fibrosis/patología , Glucosa/farmacología , Insulina/farmacología , Células Estrelladas Pancreáticas/efectos de los fármacos , Animales , Células Cultivadas , Colágeno/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Femenino , Fibrosis/metabolismo , Humanos , Ratones , Persona de Mediana Edad , Páncreas Exocrino/metabolismo , Páncreas Exocrino/patología , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Fosforilación/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo
3.
Biochem Biophys Res Commun ; 470(3): 534-538, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26801563

RESUMEN

Pancreatic islet transplantation has been recognized as an effective treatment for Type 1 diabetes; however, there is still plenty of room to improve transplantation efficiency. Because islets are metabolically active they require high oxygen to survive; thus hypoxia after transplant is one of the major causes of graft failure. Knowing the optimal oxygen tension for isolated islets would allow a transplant team to provide the best oxygen environment during pre- and post-transplant periods. To address this issue and begin to establish empirically determined guidelines for islet maintenance, we exposed in vitro cultured islets to different partial oxygen pressures (pO2) and assessed changes in islet volume, viability, metabolism, and function. Human islets were cultured for 7 days in different pO2 media corresponding to hypoxia (90 mmHg), normoxia (160 mmHg), and hyerpoxia (270 or 350 mmHg). Compared to normoxia and hypoxia, hyperoxia alleviated the loss of islet volume, maintaining higher islet viability and metabolism as measured by oxygen consumption and glucose-stimulated insulin secretion responses. We predict that maintaining pre- and post-transplanted islets in a hyperoxic environment will alleviate islet volume loss and maintain islet quality thereby improving transplant outcomes.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Técnicas de Cultivo de Órganos/métodos , Oxígeno/metabolismo , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Humanos , Secreción de Insulina
4.
J Diabetes Investig ; 13(7): 1140-1148, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35396829

RESUMEN

AIMS/INTRODUCTION: Several research groups have reported methods for quantifying pancreatic beta cell (ß-cell) injury by measuring ß-cell-specific CpG unmethylation of the insulin gene in circulation using digital droplet PCR or next-generation sequencing. However, these methods have certain disadvantages, such as the need to consider the background signal owing to the small number of target CpG sites and the need for unique equipment. MATERIALS AND METHODS: We established a novel method for detecting four CpG unmethylations of the insulin gene using two-step amplification refractory mutation system PCR. We applied it to type 1 diabetes (T1D) patients with a wide range of disease durations and to healthy adults. RESULTS: The assay showed high linearity and could detect a single copy of unmethylated insulin DNA in experiments using methylated and unmethylated plasmid DNA. The unmethylated insulin DNA level in the type 1 diabetes group, whose ß-cell mass was considerably reduced, was similar to that of healthy adults. An inverse correlation was observed between copy number and disease duration in patients with unmethylated insulin DNA-positive type 1 diabetes. CONCLUSIONS: We developed a novel method for detecting unmethylated insulin DNA in circulation that can be performed using a conventional real-time PCR system. This method would be useful for analyzing dynamic profiles of ß-cells in human disease such as type 1 diabetes.


Asunto(s)
Ácidos Nucleicos Libres de Células , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Adulto , Ácidos Nucleicos Libres de Células/metabolismo , ADN/genética , Metilación de ADN , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Mutación , Reacción en Cadena en Tiempo Real de la Polimerasa , Sulfitos
5.
Front Genet ; 11: 300, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32318096

RESUMEN

Regulatory T cells (Tregs) suppress immune responses in vivo in an antigen-specific manner. Of clinical relevance, Tregs can be isolated and expanded in vitro while maintaining immunoregulatory function. Tregs are classified as CD4+CD25highCD127low FOXP3+ cells. Demethylation of the Treg-specific demethylation region (TSDR) of FOXP3 is found in natural Tregs (nTregs). We report a method for the characterization of the differential methylation pattern of the FOXP3 TSDR in patient-derived and expanded nTregs. Human TSDR sequences from nTregs (unmethylated sequence) and pancreatic (methylated sequence) cells were amplified and cloned into plasmids. A droplet digital TaqMan probe-based qPCR (ddPCR) assay using methylation-specific primers and probes was employed to quantify unmethylated and methylated sequences. The methylation-specific droplet digital PCR (ddMSP) assay was specific and selective for unmethylated DNA in mixtures with methylated DNA in the range of 5000 copies/µL to less than 1 copy/µL (R 2 = 0.99) even in the presence of non-selective gDNAs. CD4+CD25highCD127lowFOXP3+ human nTregs, in the presence of Dynabeads or activators, were expanded for 21 days. There was a decrease in the unmethylated ratio of Tregs after expansion with essentially the same ratio at days 10, 14, and 17. However, the activator expanded group showed a significant decrease in unmethylated targets at day 21. The suppression activity of activator-expanded nTregs at day 21 was decreased compared to cells expanded with Dynabeads. These data suggest that the ddMSP can quantitatively monitor nTreg expansion in vitro. These data also indicate that the assay is sensitive and specific at differentiating nTregs from other cells and may be useful for rapid screening of nTregs in clinical protocols.

6.
Front Immunol ; 11: 712, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32411136

RESUMEN

Type 1 diabetes (T1D) arises secondary to immune-driven destruction of pancreatic ß-cells and manifests as insulin-deficient hyperglycemia. We showed that oral vaccination with live attenuated Salmonella, which simultaneously delivers autoantigens and a TGFß expression vector to immune cells in the gut mucosa, provides protection against the progression of T1D in non-obese diabetic (NOD) mice. In this study we employed the Sleeping Beauty (SB) transposon system that is composed of a transposase and transposon encoding the td-Tomato to express red fluorescent protein (RFP) to permanently mark the cells that take up the Salmonella vaccine. After animal vaccination, the transposon labeled-dendritic cells (DCs) with red fluorescence appeared throughout the secondary lymphoid tissues. Furthermore, Sleeping Beauty containing tgfß1 gene (SB-tgfß1) co-expressed TGFß and RFP. The labeled DCs were detected predominantly in Peyer's patches (PP) and mesenteric lymph nodes (MLN) and expressed CD103 surface marker. CD103+ DCs induced tolerogenic effects and gut homing. TGFß significantly increased programmed death-ligand-1 (PDL-1 or CD274) expression in the DCs in the MLN and PP of treated mice. Also, TGFß increased cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) levels in CD4+ cells in MLN and PP. Interestingly, DCs increased in all lymphatic organs of mice vaccinated with oral live Salmonella-based vaccine expressing preproinsulin (PPI), in combination with TGFß, IL10, and subtherapeutic-doses of anti-CD3 mAb compared with vehicle-treated mice. These DCs are mostly tolerogenic in MLN and PP. Furthermore the DCs obtained from vaccine-treated but not vehicle-treated mice suppressed in vitro T cell proliferation. These data suggest that the MLN and the PP are a central hub for the beneficial anti-diabetic effects of an oral Salmonella-based vaccine prevention of diabetes in rodents.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Infecciones por Salmonella/prevención & control , Vacunas contra la Salmonella/administración & dosificación , Vacunas contra la Salmonella/metabolismo , Salmonella typhimurium/inmunología , Vacunación/métodos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/metabolismo , Administración Oral , Animales , Autoantígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Plásmidos/genética , Células RAW 264.7 , Infecciones por Salmonella/microbiología , Proteína Fluorescente Roja
7.
PLoS One ; 14(8): e0221456, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31430329

RESUMEN

Gastrin is a peptide hormone, which in combination with other factors such as TGFα, EGF or GLP-1, is capable of increasing beta cell mass and lowering blood glucose levels in adult diabetic mice. In humans, administration of a bolus of gastrin alone induces insulin secretion suggesting that gastrin may target islet cells. However, whether gastrin alone is sufficient to exert an effect on isolated human islets has been controversial and the mechanism remained poorly understood. Therefore, in this study we started to examine the effects of gastrin alone on cultured adult human islets. Treatment of isolated human islets with gastrin I for 48 h resulted in increased expression of insulin, glucagon and somatostatin transcripts. These increases were significantly correlated with the levels of donor hemoglobin A1c (HbA1c) but not BMI or age. In addition, gastrin treatment resulted in increased expression of PDX1, NKX6.1, NKX2.2, MNX1 and HHEX in islets from donors with HbA1c greater than 42 mmol/mol. The addition of YM022, an antagonist of the gastrin receptor cholecystokinin B receptor (CCKBR), together with gastrin eliminated these effects, verifying that the effects of gastrin are mediated through CCKBR.CCKBR is expressed in somatostatin-expressing delta cells in islets from all donors. However, in the islets from donors with higher HbA1c (greater than 42 mmol/mol [6.0%]), cells triple-positive for CCKBR, somatostatin and insulin were detected, suggesting a de-differentiation or trans-differentiation of endocrine cells. Our results demonstrate a direct effect of gastrin on human islets from prediabetic or diabetic individuals that is mediated through CCKBR+ cells. Further, our data imply that gastrin may be a potential treatment for diabetic patients.


Asunto(s)
Gastrinas/farmacología , Hemoglobina Glucada/metabolismo , Islotes Pancreáticos/metabolismo , Donantes de Tejidos , Adolescente , Adulto , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio , Humanos , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Proteínas Nucleares , Receptor de Colecistoquinina B/metabolismo , Somatostatina/metabolismo , Factores de Transcripción , Transcripción Genética/efectos de los fármacos
8.
Front Immunol ; 10: 320, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30863412

RESUMEN

Autoimmune diseases such as type 1 diabetes (T1D) involve the loss of regulatory mechanisms resulting in increased tissue-specific cytotoxicity. The result is destruction of pancreatic insulin-producing ß-cells and loss of glucose homeostasis. We are developing a novel oral vaccine using live attenuated Salmonella to deliver TGFß, IL10, and the diabetic autoantigen preproinsulin combined with low-doses of anti-CD3 mAb. Here we show that oral administration of Salmonella-based anti-CD3 mAb combined therapy reverses new-onset T1D in non-obese diabetic (NOD) mice. The therapeutic effect of the combined therapy was associated with induction of immune suppressive CD4+CD25+Foxp3+ Treg and CD4+CD49b+LAG3+ Tr1 cells. In adoptive transfer experiments, adding or depleting Treg or Tr1 cells indicated that both are important for preventing diabetes in combined therapy-treated mice, but that Tr1 cells may have a more central role. Furthermore, induced Tr1 cells were found to be antigen-specific responding to peptide stimulation by secreting tolerance inducing IL10. These preclinical data demonstrate a role for Treg and Tr1 cells in combined therapy-mediated induction of tolerance in NOD mice. These results also demonstrate the potential of oral Salmonella-based combined therapy in the treatment of early T1D.


Asunto(s)
Traslado Adoptivo/métodos , Anticuerpos Monoclonales/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Salmonella/inmunología , Linfocitos T Reguladores/inmunología , Administración Oral , Animales , Anticuerpos Monoclonales/farmacología , Autoantígenos/genética , Autoantígenos/inmunología , Autoantígenos/metabolismo , Complejo CD3/inmunología , Terapia Combinada , Tolerancia Inmunológica/inmunología , Insulina/genética , Insulina/inmunología , Insulina/metabolismo , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos NOD , Precursores de Proteínas/genética , Precursores de Proteínas/inmunología , Precursores de Proteínas/metabolismo , Salmonella/genética , Salmonella/metabolismo , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
9.
Transplantation ; 86(2): 321-9, 2008 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-18645497

RESUMEN

BACKGROUND: The activation of p38 mitogen-activated protein kinases (MAPK) is implicated in cold ischemia-reperfusion injury of donor organs. The islet isolation process, from pancreas procurement through islet collection, may activate p38MAPK leading to cytokine release and islet damage. This damage may be prevented by treating pancreata with a p38MAPK inhibitor (p38IH) before cold preservation. METHODS: Pancreata removed from Beagle dogs were infused with University of Wisconsin solution containing the p38IH, SB203580, and Pefabloc (n=6) or vehicle (dimethyl sulfoxide and Pefabloc) alone (n=7), through the pancreatic duct and preserved using the two-layer method. After 20 to 22 hr, islets were isolated and 3000 IEQ/kg were autotransplanted into the corresponding dog to monitor glucose metabolism. RESULTS: p38IH-treated pancreata yielded significantly more islets than control pancreata (IEQ/g: 2134+/-297 vs. 1477+/-145 IEQ/g or 65,012+/-9385 vs. 45,700+/-5103 IEQ/pancreas; P<0.05). Apoptotic beta-cell percentages assessed by laser scanning cytometry were lower in p38IH-treated than the controls (44%+/-9.4% vs. 61.6%+/-4.8%, P<0.05). Tumor necrosis factor-alpha expression assessed by real-time reverse transcription polymerase chain reaction was significantly lower in the p38IH-treated group than controls. All dogs (3000 IEQ/kg) transplanted with p38IH-treated islets (n=5) became euglycemic versus four of five dogs that received untreated islets. Plasma C-peptide levels after glucagon challenge were higher in animals receiving p38IH-treated islets (n=5) versus untreated islets (n=4) (0.40+/-0.78 vs. 0.21+/-0.05 ng/mL, P<0.05). CONCLUSIONS: Infusion of pancreata with University of Wisconsin solution containing p38IH through the duct before preservation suppresses cytokine release, prevents beta-cell apoptosis, and improves islet yield significantly with no adverse effect on islet function after transplantation. p38IH treatment of human pancreata may improve islet yield for use in clinical transplantation.


Asunto(s)
Apoptosis , Inhibidores Enzimáticos/farmacología , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/patología , Preservación de Órganos/métodos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adenosina/uso terapéutico , Alopurinol/uso terapéutico , Animales , Citocinas/metabolismo , Perros , Activación Enzimática , Glutatión/uso terapéutico , Insulina/uso terapéutico , Células Secretoras de Insulina/patología , Masculino , Soluciones Preservantes de Órganos/uso terapéutico , Rafinosa/uso terapéutico , Daño por Reperfusión
10.
Vaccine ; 36(52): 8008-8018, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30416020

RESUMEN

We previously reported the development of an oral vaccine for diabetes based on live attenuated Salmonella-expressing preproinsulin (PPI) as the autoantigen. When combined with host cell-expressed TGFß, the vaccine prevented the onset of diabetes in non-obese diabetic (NOD) mice. Herein, we investigated factors that could affect vaccine efficacy including vaccination number, optimization of the autoantigen codon sequence, Salmonella SPI2-TTSS promoter/effector combinations, concurrent short-course low-dose anti-CD3. We also evaluated autoantigen GAD65 and cytokine IL10 treatment upon vaccine efficacy. T-cells we employed to elucidate the mechanism of the vaccine action. Our results showed that GAD65+TGFß or PPI+TGFß+IL10 prevented the onset of diabetes in the NOD mice and maintained glucose tolerance. However, increasing the number of vaccine doses, codon-optimization of the autoantigen(s) or use of other Salmonella promoter/effector combinations had no in vivo effect. Interestingly, two doses of vaccine (PPI+TGFß+IL10) combined with a sub-therapeutic dose of anti-CD3 prevented diabetes and decreased hyperglycemia in mice. The combined therapy also increased splenic Tregs and local Tregs in pancreatic lymph nodes (PLN) and increased regulatory (IL10 and IL2) but reduced inflammatory (IFNγ and TNFα) cytokines. Together, these results indicate that the combination of low vaccine dose number, less vaccine autoantigen expression and short-course low-dose anti-CD3 can increase regulatory mechanisms and suppress autoimmunity.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Inmunoterapia/métodos , Insulina/inmunología , Precursores de Proteínas/inmunología , Animales , Autoantígenos/administración & dosificación , Autoantígenos/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Quimioterapia Combinada , Femenino , Insulina/genética , Interleucina-10/administración & dosificación , Interleucina-10/uso terapéutico , Ratones , Ratones Endogámicos NOD , Precursores de Proteínas/genética , Salmonella , Bazo/inmunología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/administración & dosificación , Factor de Crecimiento Transformador beta/inmunología
11.
ACS Sens ; 3(1): 65-71, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29322773

RESUMEN

Label-free optical detection of insulin would allow in vitro assessment of pancreatic cell functions in their natural state and expedite diabetes-related clinical research and treatment; however, no existing method has met these criteria at physiological concentrations. Using spatially uniform 3D gold-nanoparticle sensors, we have demonstrated surface-enhanced Raman sensing of insulin in the secretions from human pancreatic islets under low and high glucose environments without the use of labels such as antibodies or aptamers. Label-free measurements of the islet secretions showed excellent correlation among the ambient glucose levels, secreted insulin concentrations, and measured Raman-emission intensities. When excited at 785 nm, plasmonic hotspots of the densely arranged 3D gold-nanoparticle pillars as well as strong interaction between sulfide linkages of the insulin molecules and the gold nanoparticles produced highly sensitive and reliable insulin measurements down to 100 pM. The sensors exhibited a dynamic range of 100 pM to 50 nM with an estimated detection limit of 35 pM, which covers the reported concentration range of insulin observed in pancreatic cell secretions. The sensitivity of this approach is approximately 4 orders of magnitude greater than previously reported results using label-free optical approaches, and it is much more cost-effective than immunoassay-based insulin detection widely used in clinics and laboratories. These promising results may open up new opportunities for insulin sensing in research and clinical applications.


Asunto(s)
Insulinas/análisis , Islotes Pancreáticos/metabolismo , Espectrometría Raman/métodos , Glucosa/farmacología , Oro , Humanos , Islotes Pancreáticos/fisiología , Nanopartículas del Metal/química , Sensibilidad y Especificidad
12.
PLoS One ; 12(10): e0185331, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28968432

RESUMEN

There is growing evidence that transplantation of cadaveric human islets is an effective therapy for type 1 diabetes. However, gauging the suitability of islet samples for clinical use remains a challenge. We hypothesized that islet quality is reflected in the expression of specific genes. Therefore, gene expression in 59 human islet preparations was analyzed and correlated with diabetes reversal after transplantation in diabetic mice. Analysis yielded 262 differentially expressed probesets, which together predict islet quality with 83% accuracy. Pathway analysis revealed that failing islet preparations activated inflammatory pathways, while functional islets showed increased regeneration pathway gene expression. Gene expression associated with apoptosis and oxygen consumption showed little overlap with each other or with the 262 probeset classifier, indicating that the three tests are measuring different aspects of islet cell biology. A subset of 36 probesets surpassed the predictive accuracy of the entire set for reversal of diabetes, and was further reduced by logistic regression to sets of 14 and 5 without losing accuracy. These genes were further validated with an independent cohort of 16 samples. We believe this limited number of gene classifiers in combination with other tests may provide complementary verification of islet quality prior to their clinical use.


Asunto(s)
Diabetes Mellitus Tipo 1/fisiopatología , Perfilación de la Expresión Génica , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Animales , Diabetes Mellitus Tipo 1/cirugía , Humanos , Modelos Logísticos , Ratones
13.
Endocrinology ; 146(1): 175-85, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15459112

RESUMEN

Silymarin is a polyphenolic flavonoid that has a strong antioxidant activity and exhibits anticarcinogenic, antiinflammatory, and cytoprotective effects. Although its hepatoprotective effect has been well documented, the effect of silymarin on pancreatic beta-cells is largely unknown. In this study, the effect of silymarin on IL-1beta and/or interferon (IFN)-gamma-induced beta-cell damage was investigated using RINm5F cells and human islets. IL-1beta and/or IFN-gamma induced cell death in a time-dependent manner in RINm5F cells. The time-dependent increase in cytokine-induced cell death appeared to correlate with the time-dependent nitric oxide (NO) production. Silymarin dose-dependently inhibited both cytokine-induced NO production and cell death in RINm5F cells. Treatment of human islets with a combination of IL-1beta and IFN-gamma (IL-1beta+IFN-gamma), for 48 h and 5 d, resulted in an increase of NO production and the impairment of glucose-stimulated insulin secretion, respectively. Silymarin prevented IL-1beta+IFN-gamma-induced NO production and beta-cell dysfunction in human islets. These cytoprotective effects of silymarin appeared to be mediated through the suppression of c-Jun NH2-terminal kinase and Janus kinase/signal transducer and activator of transcription pathways. Our data show a direct cytoprotective effect of silymarin in pancreatic beta-cells and suggest that silymarin may be therapeutically beneficial for type 1 diabetes.


Asunto(s)
Antioxidantes/farmacología , Citoprotección , Interferón gamma/farmacología , Interleucina-1/farmacología , Islotes Pancreáticos/efectos de los fármacos , Silimarina/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Combinación de Medicamentos , Activación Enzimática/efectos de los fármacos , Humanos , Interferón gamma/antagonistas & inhibidores , Interleucina-1/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas de la Leche/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/fisiología , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II , Proteínas Tirosina Quinasas/metabolismo , Ratas , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Factor de Transcripción STAT5 , Transducción de Señal/fisiología , Técnicas de Cultivo de Tejidos , Transactivadores/metabolismo
14.
Mol Immunol ; 39(13): 783-9, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12617993

RESUMEN

Methods for cell type specific targeted intracellular delivery of proteins in vivo remain limited. A murine monoclonal anti-dsDNA antibody, mAb 3E10, was selectively transported into skeletal muscle cells in vivo. The antibody bound a 200 kDa protein only found in lysates of skeletal muscle by Western blotting. The 200 kDa protein was purified from muscle lysate by antibody affinity chromatography and identified as the skeletal muscle specific heavy chain of myosin IIb by electrospray mass spectrometry. Antibody binding specificity for myosin IIb was demonstrated in Western blots by binding myosin in skeletal muscle lysates from mice null for myosin IId but not in mice null for myosin IIb. Myosin IIb is implicated in the specific targeting of mAb 3E10 to skeletal muscle.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Músculo Esquelético/inmunología , Miosina Tipo IIB no Muscular/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antinucleares/administración & dosificación , Anticuerpos Antinucleares/metabolismo , Anticuerpos Monoclonales/farmacocinética , Especificidad de Anticuerpos , Femenino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Miosina Tipo IIB no Muscular/deficiencia , Miosina Tipo IIB no Muscular/genética , Miosina Tipo IIB no Muscular/metabolismo , Ratas , Distribución Tisular
15.
Transplant Direct ; 1(4)2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26146662

RESUMEN

BACKGROUND: We evaluated three commercially available enzymes for pancreatic digestion by comparing key parameters during the islet isolation process, as well as islet quality post-isolation. METHODS: Retrospectively compared and analyzed islet isolations from pancreata using three different enzyme groups: Liberase HI (n=63), Collagenase NB1/Neutral Protease (NP) (n=43), and Liberase Mammalian Tissue Free Collagenase/Thermolysin (MTF C/T) (n=115). A standardized islet isolation and purification method was used. Islet quality assessment was carried out using islet count, viability, in vitro glucose-stimulated insulin secretion (GSIS), glucose-stimulated oxygen consumption rate (ΔOCR), and in vivo transplantation model in mice. RESULTS: Donor characteristics were not significantly different among the three enzyme groups used in terms of age, sex, hospital stay duration, cause of death, body mass index (BMI), hemoglobin A1c (HbA1c), cold ischemia time (CIT), and pancreas weight. Digestion efficacy (percentage of digested tissue by weight) was significantly higher in the Liberase MTF C/T group (73.5 ± 1.5 %) when compared to the Liberase HI group (63.6 ± 2.3 %) (p<0.001) and the Collagenase NB1/NP group (61.7 ± 2.9%) (p<0.001). The stimulation index for GSIS was significantly higher in the Liberase MTF C/T group (5.3 ± 0.5) as compared to the Liberase HI (2.9 ± 0.2) (p<0.0001) and the Collagenase NB1/NP (3.6 ± 2.9) (p=0.012) groups. Furthermore, the Liberase MTF C/T enzymes showed the highest success rate of transplantation in diabetic NOD Scid mice (65%), which was significantly higher than the Liberase HI (42%, p=0.001) and the Collagenase NB1/NP enzymes (41%, p<0.001). CONCLUSIONS: Liberase MTF C/T is superior to Liberase HI and Collagenase NB1/NP in terms of digestion efficacy and glucose-stimulated insulin secretion in vitro. Moreover, Liberase MTF C/T had a significantly higher success rate of transplantation in diabetic NOD Scid mice compared to Liberase HI and Collagenase NB1/NP enzymes.

16.
Cell Transplant ; 24(9): 1879-86, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25198342

RESUMEN

UNLABELLED: The aim of this study was to investigate the effects of elevated donor HbA1c levels (type 2 diabetes, T2D) on the islet yield and functionality postisolation. In this retrospective analysis, donors for islet isolations were classified into two groups: T2D group (HbA1c ≥ 6.5%, n = 18) and normal group (HbA1c < 6.5%, n = 308). Optimum pancreas digestion time (switch time) was significantly higher in the T2D group compared to the normal group (13.7 ± 1.2 vs. 11.7 ± 0.1 min, respectively, p = 0.005). Islet yields were significantly lower in the T2D group compared to the control (T2D vs. control): islet equivalent (IEQ)/g (prepurification 2,318 ± 195 vs. 3,713 ± 114, p = 0.003; postpurification 1,735 ± 175 vs. 2,663 ± 89, p = 0.013) and islet particle number (IPN)/g (prepurification, 2,519 ± 336 vs. 4,433 ± 143, p = 0.001; postpurification, 1,760 ± 229 vs. 2,715 ± 85, p = 0.007). Islets from T2D pancreata had significantly lower viability (T2D vs. CONTROL: 91.9 ± 1.6 vs. 94.4 ± 0.3%, p = 0.004) and decreased oxygen consumption rate (ΔOCR) (T2D vs. CONTROL: 0.09 ± 0.01 and 0.21 ± 0.03 nmol O2 100 islets(-1) min(-1), p = 0.049). The islets isolated from T2D donor pancreata reversed diabetes in NOD-SCID mice in 9% (2/22) compared to islets from control donor pancreata, which reversed diabetes in 67% (175/260, p < 0.001). In conclusion, this study demonstrates that elevated HbA1c (≥ 6.5%) is associated with impairment of islet function and lower islet yield; however, these islets could not be suitable for clinical applications.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Hemoglobina Glucada/análisis , Islotes Pancreáticos/citología , Adulto , Animales , Supervivencia Celular , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Glucosa/farmacología , Humanos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Consumo de Oxígeno/efectos de los fármacos , Donantes de Tejidos , Trasplante Heterólogo
17.
Cancer Lett ; 195(2): 211-9, 2003 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-12767530

RESUMEN

scFv fragments of a monoclonal antibody that penetrates living cells and localizes in nuclei were designed as fusion proteins with C-terminal p53 peptides and tested for restoring p53 function in p53 mutant cancer cells. scFv fragments transported a 30-mer C-terminal peptide of p53 into cancer cells and induced cellular cytotoxicity in contrast to scFv fragments alone and other scFv-p53 fusion peptides. Cellular toxicity was not observed with scFv fragments containing a single mutation in VH that prevented antibody penetration. Our results demonstrate the potential efficacy of antibody scFv fragments as a nuclear delivery system in cancer cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Núcleo Celular/metabolismo , Inmunoconjugados/administración & dosificación , Región Variable de Inmunoglobulina/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Activación Transcripcional/efectos de los fármacos , Proteína p53 Supresora de Tumor/administración & dosificación , Transporte Activo de Núcleo Celular , Adenocarcinoma/patología , Animales , Anticuerpos Monoclonales/administración & dosificación , Neoplasias Óseas/patología , Células CHO , Células COS , Permeabilidad de la Membrana Celular , Chlorocebus aethiops , Neoplasias Colorrectales/patología , Cricetinae , Sistemas de Liberación de Medicamentos , Genes Reporteros , Humanos , Inmunoconjugados/farmacología , Inmunoconjugados/toxicidad , Región Variable de Inmunoglobulina/farmacología , Ratones , Ratones Endogámicos MRL lpr , Osteosarcoma/patología , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/toxicidad , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacología , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/fisiología , Proteína p53 Supresora de Tumor/toxicidad
18.
PLoS One ; 9(4): e94591, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24722187

RESUMEN

The onset of metabolic dysregulation in type 1 diabetes (T1D) occurs after autoimmune destruction of the majority of pancreatic insulin-producing beta cells. We previously demonstrated that the DNA encoding the insulin gene is uniquely unmethylated in these cells and then developed a methylation-specific PCR (MSP) assay to identify circulating beta cell DNA in streptozotocin-treated mice prior to the rise in blood glucose. The current study extends to autoimmune non-obese diabetic (NOD) mice and humans, showing in NOD mice that beta cell death occurs six weeks before the rise in blood sugar and coincides with the onset of islet infiltration by immune cells, demonstrating the utility of MSP for monitoring T1D. We previously reported unique patterns of methylation of the human insulin gene, and now extend this to other human tissues. The methylation patterns of the human insulin promoter, intron 1, exon 2, and intron 2 were determined in several normal human tissues. Similar to our previous report, the human insulin promoter was unmethylated in beta cells, but methylated in all other tissues tested. In contrast, intron 1, exon 2 and intron 2 did not exhibit any tissue-specific DNA methylation pattern. Subsequently, a human MSP assay was developed based on the methylation pattern of the insulin promoter and human islet DNA was successfully detected in circulation of T1D patients after islet transplantation therapy. Signal levels of normal controls and pre-transplant samples were shown to be similar, but increased dramatically after islet transplantation. In plasma the signal declines with time but in whole blood remains elevated for at least two weeks, indicating that association of beta cell DNA with blood cells prolongs the signal. This assay provides an effective method to monitor beta cell destruction in early T1D and in islet transplantation therapy.


Asunto(s)
Muerte Celular/fisiología , Metilación de ADN , Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/metabolismo , Insulina/genética , Reacción en Cadena de la Polimerasa/métodos , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Humanos , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos NOD
19.
Vaccine ; 32(20): 2300-7, 2014 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-24631074

RESUMEN

Type 1 diabetes (T1D) is a metabolic disease that is initiated by the autoimmune destruction of pancreatic insulin-producing beta cells that is accompanied by the development of antigen-specific antibodies and cytotoxic T lymphocytes (CTLs). Several studies have shown that vaccination with diabetic autoantigens provides some protection against this process. In this report we describe a new oral vaccine that utilizes live attenuated Salmonella for simultaneous delivery of autoantigens in conjunction with immunomodulatory cytokine genes to immune cells in the gut mucosa. Recent data showed that live attenuated Salmonella is a safe, simple and effective vector for expression of antigens and cytokines by antigen-presenting cells (APCs) of gut-associated lymphatic tissue (GALT). This novel strategy was tested by fusion of the diabetic autoantigen preproinsulin with Salmonella secretory effector protein (SseF) of pathogenicity island-2 (SPI2). In this way the autoantigen is only expressed inside the host immune cells and translocated to the host cell cytosol. In addition Salmonella was used to deliver the gene for the immunomodulatory cytokine transforming growth factor beta (TGFß) for host cell expression. Oral co-vaccination of 8 week-old non-obese diabetic (NOD) mice with three weekly doses of both the autoantigen and cytokine significantly reduced the development of diabetes, improved the response to glucose challenge, preserved beta cell mass, and reduced the severity of insulitis compared with controls and autoantigen alone. Combination therapy also resulted in increased circulating levels of IL10 four weeks post-vaccination and IL2 for 12 weeks post-vaccination, but without effect on proinflammatory cytokines IL6, IL12(p70), IL17 and IFNγ. However, in non-responders there was a significant rise in IL12 compared with responders. Future studies will examine the mechanism of this vaccination strategy in more detail. In conclusion, Salmonella-based oral vaccines expressing autoantigens combined with imunomodulatory cytokines appears to be a promising therapy for prevention of T1D.


Asunto(s)
Autoantígenos/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Insulina/inmunología , Precursores de Proteínas/inmunología , Salmonella , Vacunas/inmunología , Administración Oral , Animales , Autoantígenos/genética , Proteínas Bacterianas/inmunología , Línea Celular , Femenino , Vectores Genéticos , Insulina/genética , Interleucinas/inmunología , Proteínas de la Membrana/inmunología , Ratones Endogámicos NOD , Precursores de Proteínas/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
20.
PLoS One ; 7(10): e47942, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144715

RESUMEN

DNA methylation is a mechanism by which cells control gene expression, and cell-specific genes often exhibit unique patterns of DNA methylation. We previously reported that the mouse insulin-2 gene (Ins2) promoter has three potential methylation (CpG) sites, all of which are unmethylated in insulin-producing cells but methylated in other tissues. In this study we examined Ins2 exon 2 and found a similar tissue-specific methylation pattern. These methylation patterns can differentiate between DNA from insulin-producing beta cells and other tissues. We hypothesized that damaged beta cells release their DNA into circulation at the onset of type 1 diabetes mellitus (T1DM) and sought to develop a quantitative methylation-specific polymerase chain reaction (qMSP) assay for circulating beta cell DNA to monitor the loss of beta cells. Methylation-specific primers were designed to interrogate two or more CpG in the same assay. The cloned mouse Ins2 gene was methylated in vitro and used for development of the qMSP assay. We found the qMSP method to be sensitive and specific to differentiate between insulin-producing cells and other tissues with a detection limit of 10 copies in the presence of non-specific genomic DNA background. We also compared different methods for data analysis and found that the Relative Expression Ratio method is the most robust method since it incorporates both a reference value to normalize day-to-day variability as well as PCR reaction efficiencies to normalize between the methylation-specific and bisulfite-specific components of the calculations. The assay was applied in the streptozotocin-treated diabetic mouse model and detected a significant increase in circulating beta cell DNA before the rise in blood glucose level. These results demonstrate that this qMSP assay can be used for monitoring circulating DNA from insulin-producing cells, which will provide the basis for development of assays to detect beta cell destruction in early T1DM.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Animales , Secuencia de Bases , Muerte Celular/genética , ADN/sangre , ADN/genética , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/patología , Exones/genética , Insulina/genética , Células Secretoras de Insulina/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Datos de Secuencia Molecular , Reproducibilidad de los Resultados , Homología de Secuencia de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA