Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros










Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1089817, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36875761

RESUMEN

Type 1 diabetes mellitus (T1DM) is an autoimmune disorder specifically targeting pancreatic islet beta cells. Despite many efforts focused on identifying new therapies able to counteract this autoimmune attack and/or stimulate beta cells regeneration, TD1M remains without effective clinical treatments providing no clear advantages over the conventional treatment with insulin. We previously postulated that both the inflammatory and immune responses and beta cell survival/regeneration must be simultaneously targeted to blunt the progression of disease. Umbilical cord-derived mesenchymal stromal cells (UC-MSC) exhibit anti-inflammatory, trophic, immunomodulatory and regenerative properties and have shown some beneficial yet controversial effects in clinical trials for T1DM. In order to clarify conflicting results, we herein dissected the cellular and molecular events derived from UC-MSC intraperitoneal administration (i.p.) in the RIP-B7.1 mouse model of experimental autoimmune diabetes. Intraperitoneal (i.p.) transplantation of heterologous mouse UC-MSC delayed the onset of diabetes in RIP-B7.1 mice. Importantly, UC-MSC i. p. transplantation led to a strong peritoneal recruitment of myeloid-derived suppressor cells (MDSC) followed by multiple T-, B- and myeloid cells immunosuppressive responses in peritoneal fluid cells, spleen, pancreatic lymph nodes and the pancreas, which displayed significantly reduced insulitis and pancreatic infiltration of T and B Cells and pro-inflammatory macrophages. Altogether, these results suggest that UC-MSC i. p. transplantation can block or delay the development of hyperglycemia through suppression of inflammation and the immune attack.

2.
Trials ; 22(1): 674, 2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34600562

RESUMEN

BACKGROUND: Moderate/severe cases of COVID-19 present a dysregulated immune system with T cell lymphopenia and a hyper-inflammatory state. This is a study protocol of an open-label, multi-center, double-arm, randomized, dose-finding phase I/II clinical trial to evaluate the safety, tolerability, alloreactivity, and efficacy of the administration of allogeneic memory T cells and natural killer (NK) cells in COVID-19 patients with lymphopenia and/or pneumonia. The aim of the study is to determine the safety and the efficacy of the recommended phase 2 dose (RP2D) of this treatment for patients with moderate/severe COVID-19. METHODS: In the phase I trial, 18 patients with COVID-19-related pneumonia and/or lymphopenia with no oxygen requirement or with an oxygen need of ≤ 2.5 liters per minute (lpm) in nasal cannula will be assigned to two arms, based on the biology of the donor and the patient. Treatment of arm A consists of the administration of escalating doses of memory T cells, plus standard of care (SoC). Treatment of arm B consists of the administration of escalating doses of NK cells, plus SoC. In the phase II trial, a total of 182 patients with COVID-19-related pneumonia and/or lymphopenia requiring or not oxygen supplementation but without mechanical ventilation will be allocated to arm A or B, considering HLA typing. Within each arm, they will be randomized in a 1:1 ratio. In arm A, patients will receive SoC or RP2D for memory T cells plus the SoC. In arm B, patients will receive SoC or RP2D for NK cells plus the SoC. DISCUSSION: We hypothesized that SARS-CoV-2-specific memory T-lymphocytes obtained from convalescent donors recovered from COVID-19 can be used as a passive cell immunotherapy to treat pneumonia and lymphopenia in moderate/severe patients. The lymphopenia induced by COVID-19 constitutes a therapeutic window that may facilitate donor engraftment and viral protection until recovery. TRIAL REGISTRATION: ClinicalTrials.gov NCT04578210 . First Posted : October 8, 2020.


Asunto(s)
COVID-19 , Linfopenia , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Humanos , Memoria Inmunológica , Células Asesinas Naturales , Linfopenia/diagnóstico , Linfopenia/terapia , Estudios Multicéntricos como Asunto , Ensayos Clínicos Controlados Aleatorios como Asunto , SARS-CoV-2 , Linfocitos T , Resultado del Tratamiento
3.
EClinicalMedicine ; 39: 101086, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34405140

RESUMEN

BACKGROUND: Effective treatments are still needed to reduce the severity of symptoms, time of hospitalization, and mortality of COVID-19. SARS-CoV-2 specific memory T-lymphocytes obtained from convalescent donors recovered can be used as passive cell immunotherapy. METHODS: Between September and November 2020 a phase 1, dose-escalation, single centre clinical trial was conducted to evaluate the safety and feasibility of the infusion of CD45RA- memory T cells containing SARS-CoV-2 specific T cells as adoptive cell therapy against moderate/severe cases of COVID-19. Nine participants with pneumonia and/or lymphopenia and with at least one human leukocyte antigen (HLA) match with the donor were infused. The first three subjects received the lowest dose (1 × 105 cells/kg), the next three received the intermediate dose (5 × 105 cells/kg) and the last three received the highest dose (1 × 106 cells/kg) of CD45RA- memory T cells. Clinicaltrials.gov registration: NCT04578210. FINDINGS: All participants' clinical status measured by National Early Warning Score (NEWS) and 7-category point ordinal scales showed improvement six days after infusion. No serious adverse events were reported. Inflammatory parameters were stabilised post-infusion and the participants showed lymphocyte recovery two weeks after the procedure. Donor microchimerism was observed at least for three weeks after infusion in all patients. INTERPRETATION: This study provides preliminary evidence supporting the idea that treatment of COVID-19 patients with moderate/severe symptoms using convalescent CD45RA- memory T cells is feasible and safe. FUNDING: Clinical Trial supported by Spanish Clinical Research Network PT17/0017/0013. Co-funded by European Regional Development Fund/European Social Fund. CRIS CANCER Foundation Grant to AP-M and Agencia Valenciana de Innovación Grant AVI-GVA COVID-19-68 to BS.

4.
Front Cell Dev Biol ; 9: 620730, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33718360

RESUMEN

Syndrome coronavirus 2 (SARS-CoV-2) pandemic is causing a second outbreak significantly delaying the hope for the virus' complete eradication. In the absence of effective vaccines, we need effective treatments with low adverse effects that can treat hospitalized patients with COVID-19 disease. In this study, we determined the existence of SARS-CoV-2-specific T cells within CD45RA- memory T cells in the blood of convalescent donors. Memory T cells can respond quickly to infection and provide long-term immune protection to reduce the severity of COVID-19 symptoms. Also, CD45RA- memory T cells confer protection from other pathogens encountered by the donors throughout their life. It is of vital importance to resolve other secondary infections that usually develop in patients hospitalized with COVID-19. We found SARS-CoV-2-specific memory T cells in all of the CD45RA- subsets (CD3+, CD4+, and CD8+) and in the central memory and effector memory subpopulations. The procedure for obtaining these cells is feasible, easy to implement for small-scale manufacture, quick and cost-effective, involves minimal manipulation, and has no GMP requirements. This biobank of specific SARS-CoV-2 memory T cells would be immediately available "off-the-shelf" to treat moderate/severe cases of COVID-19, thereby increasing the therapeutic options available for these patients.

5.
Stem Cell Res ; 16(3): 568-78, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26999760

RESUMEN

Little is known about the functions of downstream regulatory element antagonist modulator (DREAM) in embryonic stem cells (ESCs). However, DREAM interacts with cAMP response element-binding protein (CREB) in a Ca(2+)-dependent manner, preventing CREB binding protein (CBP) recruitment. Furthermore, CREB and CBP are involved in maintaining ESC self-renewal and pluripotency. However, a previous knockout study revealed the protective function of DREAM depletion in brain aging degeneration and that aging is accompanied by a progressive decline in stem cells (SCs) function. Interestingly, we found that DREAM is expressed in different cell types, including human ESCs (hESCs), human adipose-derived stromal cells (hASCs), human bone marrow-derived stromal cells (hBMSCs), and human newborn foreskin fibroblasts (hFFs), and that transitory inhibition of DREAM in hESCs reduces their pluripotency, increasing differentiation. We stipulate that these changes are partly mediated by increased CREB transcriptional activity. Overall, our data indicates that DREAM acts in the regulation of hESC pluripotency and could be a target to promote or prevent differentiation in embryonic cells.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , Proteínas de Interacción con los Canales Kv/metabolismo , Tejido Adiposo/citología , Antígenos de Superficie/metabolismo , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Diferenciación Celular , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación hacia Abajo , Fibroblastos/citología , Fibroblastos/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Proteínas de Interacción con los Canales Kv/antagonistas & inhibidores , Proteínas de Interacción con los Canales Kv/genética , Proteoglicanos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Antígenos Embrionarios Específico de Estadio/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
6.
Cell Death Dis ; 5: e1304, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24967966

RESUMEN

Recent studies suggested that the post-natal mesothelium retain differentiative potential of the embryonic mesothelium, which generates fibroblasts and vascular smooth muscle cells (VSMCs), in developing coelomic organs via epithelial-to-mesenchymal transition (EMT). Whether adult mesothelial cells (MCs) are able to give rise to functional VSMCs in vitro and which are the factors and mechanisms directing this process remain largely unknown. Here, we isolated adipose tissue MCs (ATMCs) from adult mice, and demonstrated that ATMCs cultured in a serum-containing media supplemented with epidermal growth factor (EGF) efficiently increased both their proliferation and EMT above levels found in only serum-containing media cultures. EGF-induced ATMCs gained phosphorylation of the EGF receptor and activated simultaneously ILK/Erk1/2, PI3K/Akt and Smad2/3-dependent pathways. Sequential subculture onto collagen-I surface efficiently improved their vasculogenic EMT towards cells featuring VSMCs (α-SMA, calponin, caldesmon, SM22α, desmin, SM-MHC, smoothelin-B and PDGFR-ß) that could actively contract in response to receptor and non-receptor-mediated vasoactive agonists. Overall, our results indentify EGF signalling as a robust vasculogenic inductive pathway for ATMCs, leading to their transdifferentiation into functional VSMC-like cells.


Asunto(s)
Tejido Adiposo/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Tejido Adiposo/citología , Animales , Antígenos de Diferenciación/metabolismo , Células Cultivadas , Femenino , Ratones , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Ratas , Ratas Wistar , Transducción de Señal/fisiología
7.
Cell Death Dis ; 4: e570, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23559004

RESUMEN

Lineage commitment during embryonic stem cell (ESC) differentiation is controlled not only by a gamut of transcription factors but also by epigenetic events, mainly histone deacetylation and promoter DNA methylation. The DNA demethylation agent 5'-aza-2'-deoxycytidine (AzadC) has been widely described as an effective promoter of cardiomyogenic differentiation in various stem cell types. However, its toxicity and instability complicate its use. Therefore, the purpose of this study was to examine the effects of zebularine (1-(ß-D-ribofuranosyl)-1,2-dihydropyrimidin-2-1), a stable and non-toxic DNA cytosine methylation inhibitor, on mouse ESC (mESC) differentiation. Herein, we report that treating embryoid bodies, generated from mESCs, with 30 µM zebularine for 7 days led to greater cell differentiation and induced the expression of several cardiac-specific markers that were detected using reverse transcription-polymerase chain reaction (RT-PCR), real-time PCR, immunostaining and flow cytometry. Zebularine enhanced the expression of cardiac markers and the appearance of beating cells that responded to cardiac drugs, including ion channel blockers (diltiazem) and ß-adrenergic stimulators (isoproterenol). Gene promoter methylation status was assessed using methylation-specific PCR (MSP) and validated by bisulfite sequencing analysis. Global gene expression profiling using microarrays showed that zebularine-differentiated cells are distinct from control ESCs. Pathway analysis revealed an enhancement of cellular processes such as embryonic development, cardiovascular system development and function. In addition, the whole-cell proteins exhibited different profiles as analyzed by two-dimensional differential-in-gel-electrophoresis. Our results indicate that zebularine regulates mesodermal differentiation of mESCs, controls promoter methylation of crucial cardiac genes and may help to improve cardiomyogenic differentiation.


Asunto(s)
Citidina/análogos & derivados , Cuerpos Embrioides/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Citidina/farmacología , Metilación de ADN/efectos de los fármacos , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Cell Death Differ ; 17(6): 1025-33, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20075941

RESUMEN

Exposure of mouse embryonic stem (mES) cells to high concentrations of chemical nitric oxide (NO) donors promotes differentiation, but the mechanisms involved in this process at the gene expression level are poorly defined. In this study we report that culture of mES cells in the presence of 0.25-1.0 mM diethylenetriamine nitric oxide adduct (DETA-NO) leads to downregulation of Nanog and Oct4, the two master genes involved in the control of the pluripotent state. This action of NO was also apparent in the human ES cell line, HS 181. The suppressive action of NO on Nanog gene depends on the activation of p53 repressor protein by covalent modifications, such as pSer15, pSer315, pSer392 and acetyl Lys 379. NO-induced repression of Nanog is also associated with binding of trimethylated histone H3 and pSer315 p53 to its promoter region. In addition, exposure to 0.5 mM DETA-NO induces early differentiation events of cells with acquisition of epithelial morphology and expression of markers of definitive endoderm, such as FoxA2, Gata4, Hfn1-beta and Sox 17. This phenotype was increased when cells were treated with valproic acid (VPA) for 10 days.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/genética , Óxido Nítrico/metabolismo , Animales , Apoptosis , Diferenciación Celular , Línea Celular , Regulación hacia Abajo , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Endodermo/citología , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Proteína Homeótica Nanog , Donantes de Óxido Nítrico/farmacología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Proteína p53 Supresora de Tumor/metabolismo
9.
Cell Death Dis ; 1: e80, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21368853

RESUMEN

Nitric oxide (NO) is an intracellular messenger in several cell systems, but its contribution to embryonic stem cell (ESC) biology has not been characterized. Exposure of ESCs to low concentrations (2-20 µM) of the NO donor diethylenetriamine NO adduct confers protection from apoptosis elicited by leukaemia inhibitory factor (LIF) withdrawal. NO blocked caspase 3 activation, PARP degradation, downregulation of the pro-apoptotic genes Casp7, Casp9, Bax and Bak1 and upregulation of the anti-apoptotic genes Bcl-2 111, Bcl-2 and Birc6. These effects were also observed in cells overexpressing eNOS. Exposure of LIF-deprived mESCs to low NO prevented the loss of expression of self-renewal genes (Oct4, Nanog and Sox2) and the SSEA marker. Moreover, NO blocked the differentiation process promoted by the absence of LIF and bFGF in mouse and human ESCs. NO treatment decreased the expression of differentiation markers, such as Brachyury, Gata6 and Gata4. Constitutive overexpression of eNOS in cells exposed to LIF deprivation maintained the expression of self-renewal markers, whereas the differentiation genes were repressed. These effects were reversed by addition of the NOS inhibitor L-NMMA. Altogether, the data suggest that low NO has a role in the regulation of ESC differentiation by delaying the entry into differentiation, arresting the loss of self-renewal markers and promoting cell survival by inhibiting apoptosis.


Asunto(s)
Células Madre Embrionarias/citología , Óxido Nítrico/metabolismo , Animales , Apoptosis , Diferenciación Celular , Supervivencia Celular , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Factor Inhibidor de Leucemia/metabolismo , Antígeno Lewis X/metabolismo , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Poliaminas/farmacología , Factores de Transcripción SOXB1/metabolismo
10.
Cells Tissues Organs ; 188(1-2): 70-7, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18305378

RESUMEN

Diabetes is a chronic disease characterized by a deficit in beta cell mass and a failure of glucose homeostasis. Both circumstances result in a variety of severe complications and an overall shortened life expectancy. Thus, diabetes represents an attractive candidate for cell therapy. Reversal of diabetes can be achieved through pancreas and islet transplantation, but shortage of donor organs has prompted an intensive search for alternative sources of beta cells. This achievement has stimulated the search for appropriate stem cell sources. Both embryonic and adult stem cells have been used to generate surrogate beta cells or otherwise restore beta cell functioning. In this regard, several studies have reported the generation of insulin-secreting cells from embryonic and adult stem cells that normalized blood glucose values when transplanted into diabetic animal models. Due to beta cell complexity, insulin-producing cells generated from stem cells do not possess all beta cell attributes. This indicates the need for further development of methods for differentiation and selection of completely functional beta cells. While these problems are overcome, diabetic patients may benefit from therapeutic strategies based on autologous stem cell therapies addressing late diabetic complications. In this article, we discuss the recent progress in the generation of insulin-producing cells from embryonic and adult stem cells, together with the challenges for the clinical use of diabetes stem cell therapy.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Diabetes Mellitus/terapia , Células Madre/citología , Células Madre Adultas/citología , Linaje de la Célula , Complicaciones de la Diabetes/terapia , Células Madre Embrionarias/citología , Humanos , Células Secretoras de Insulina/citología , Donantes de Tejidos
11.
J. physiol. biochem ; 62(4): 253-262, dic. 2006.
Artículo en Inglés | IBECS | ID: ibc-122987

RESUMEN

The capability of halocin H6 (a bacteriocin-like protein produced by haloarchaeaHaloferax gibbonsii) to inhibit Na+/H+ exchange (NHE) in mammalian cells and its cardio-protective efficacy on the ischemic and reperfused myocardium were evaluated in the present study. H6 inhibits NHE activity (measured by a flow cytometry method) in a dose-dependent form of cell lines of mammalian origin (HEK293, NIH3T3, Jurkat and HL-1) as well as in primary cell culture from human skeletal muscle (myocytes and fibroblasts).In vivo, an ischemia-reperfusion model in dogs by coronary arterial occlusion was used (two hours of regional ischemia and three hours of reperfusion). In animals treated with halocin H6 there was a significant reduction of premature ventricular ectopic beats and infarct size, whereas blood pressure and heart rate remained unchanged. Up to date, halocin H6 is the only described biological molecule that exerts a, specific inhibitory activity in NHE of eukaryotic cells (AU)


En el presente trabajo se evalúa la capacidad de la halocina H6 (una proteína tipo bacteriocina producida por la haloarchaeaHaloferax gibbonsii) para inhibir el intercambiador Na+/H+ (NHE) de céludas de mamífero y su posible eficacia cardioprotectora frente a los daños causados por isquemia-reperfusión del miocardio. En experimentosin vitro H6 inhibe la actividad de NHE (determinada por citometría de flujo) de forma dosis-dependiente tanto en líneas celulares de mamíferos (HEK293, NIH3T3, Jurkat y HL-1) como en cultivos primarios de miocitos y fibroblastos aislados de músculo esquelético humano. En experimentosin vivo se utilizó un modelo de isquemia-reperfusión en perros por oclusión de la arteria coronaria (dos horas de isquemia y tres de reperfusión). En animales tratados con halocina H6 se produjo una disminución significativa a nivel estadístico, tanto del número de latidos ectópicos ventriculares como del tamaño del infarto, mientras que no se produjeron cambios tanto en la presión sanguínea como en el ritmo cardíaco. Hasta la fecha la halocina H6 es la única molécula biológica descrita que ejerce una actividad inhibidora específica sobre el NHE de células eucariotas (AU)


Asunto(s)
Animales , Perros , /antagonistas & inhibidores , Bacteriocinas/farmacocinética , Cardiotónicos/farmacocinética , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Sustancias Protectoras/farmacocinética , Modelos Animales de Enfermedad
13.
Handb Exp Pharmacol ; (174): 147-67, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16370328

RESUMEN

Stem cells possess the ability to self-renew by symmetric divisions and, under certain circumstances, differentiate to a committed lineage by asymmetric cell divisions. Depending on the origin, stem cells are classified as either embryonic or adult. Embryonic stem cells are obtained from the inner cell mass of the blastocyst, a structure that appears during embryonic development at day 6 in humans and day 3.5 in mice. Adult stem cells are present within tissues of adult organisms and are responsible for cell turnover or repopulation of tissues under normal or exceptional circumstances. Taken together, stem cells might represent a potential source of tissues for cell therapy protocols, and diabetes is a candidate disease that may benefit from cell replacement protocols. The pathology of type 1 diabetes is caused by the autoimmune destruction or malfunction of pancreatic beta cells, and consequently, a lack of insulin. The absence of insulin is life-threatening, thus requiring diabetic patients to take daily hormone injections from exogenous sources; however, insulin injections do not adequately mimic beta cell function. This results in the development of diabetic complications such as neuropathy, nephropathy, retinopathy and diverse cardiovascular disorders. This chapter intends to summarize the possibilities opened by embryonic and adult stem cells in regenerative medicine for the cure of diabetes.


Asunto(s)
Diabetes Mellitus/patología , Diabetes Mellitus/terapia , Trasplante de Islotes Pancreáticos , Células Madre/citología , Células Madre/metabolismo , Animales , Diferenciación Celular , Embrión de Mamíferos/citología , Humanos , Páncreas/crecimiento & desarrollo
14.
J Physiol Biochem ; 62(4): 253-62, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17615951

RESUMEN

The capability of halocin H6 (a bacteriocin-like protein produced by haloarchaea Haloferax gibbonsii) to inhibit Na+/H+ exchanger (NHE) in mammalian cells and its cardio-protective efficacy on the ischemic and reperfused myocardium were evaluated in the present study. H6 inhibits NHE activity (measured by a flow cytometry method) in a dose-dependent form of cell lines of mammalian origin (HEK293, NIH3T3, Jurkat and HL-1) as well as in primary cell culture from human skeletal muscle (myocytes and fibroblasts). In vivo, an ischemia-reperfusion model in dogs by coronary arterial occlusion was used (two hours of regional ischemia and three hours of reperfusion). In animals treated with halocin H6 there was a significant reduction of premature ventricular ectopic beats and infarct size, whereas blood pressure and heart rate remained unchanged. Up to date, halocin H6 is the only described biological molecule that exerts a specific inhibitory activity in NHE of eukaryotic cells.


Asunto(s)
Archaea/química , Bacteriocinas/farmacología , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Animales , Bacteriocinas/aislamiento & purificación , Línea Celular , Humanos , Ratones
15.
Transplant Proc ; 37(8): 3404-6, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16298608

RESUMEN

UNLABELLED: Islet transplantation is a promising therapy in the treatment of diabetes mellitus. Herein we present the result from the first series of islet isolations carried out in our new islet isolation facility. The aims of study were to analyze the influence of various donor characteristics on the success of islet isolation and compare these outcomes with other European and American groups. Data from 22 completed islet isolation were used to compare donor and isolation variables among successful (>300,000 IEQs) versus unsuccessful isolations. The successful isolation rate from our laboratory was 31.8%. We did not see any significant differences between successful and unsuccessful groups according to donor characteristics, although age was close to significance (38.57 +/- 10.29 versus 48.33 +/- 12.39; P = .08). Donor age (1.12 [1.23; 0.99]) and body mass index (0.065 [1.32; 3.08]) were associated with isolation success in a logistic regression model. We did not find differences among intraprocedure variables with the exception of IEQ prepurification (409,073 +/- 115,041 versus 263,776 +/- 128,988; P < .05). IEQpre and IEQpost were positively correlated (P < .05). In comparison with other groups, we observed differences in some cases related to islet yield prepurification (P < .05) but not postpurification. Purity from our islet preparations was the highest from all considered groups (P < .05). Recovery was similar in all groups. CONCLUSIONS: In our experience, donor characteristics have no influence on the success rate. The digestion step is a critical factor for success. Our results with respect to IE yield were close to that of experienced groups.


Asunto(s)
Islotes Pancreáticos/citología , Recolección de Tejidos y Órganos/normas , Adulto , Índice de Masa Corporal , Cadáver , Separación Celular/métodos , Humanos , Persona de Mediana Edad , Análisis de Regresión , España , Donantes de Tejidos , Recolección de Tejidos y Órganos/métodos
16.
Curr Med Chem ; 11(20): 2707-16, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15544471

RESUMEN

Diabetes Mellitus is by far one of the most propagated chronic diseases, affecting 150 million people worldwide. This affliction is caused by a malfunction of pancreatic endocrine cells, which provokes a failure in the insulin release and glucose homeostasis. Plasma membrane K(ATP) channels have a key role in the stimulus-secretion coupling of pancreatic beta-cells. Consequently, many investigations have developed efficient drugs for the treatment of diabetes, such as sulphonylureas, which specifically close K(ATP) channels leading to an enhanced insulin secretion. Recent studies show that, in addition to its well-known plasma membrane location, sulphonylurea receptors and sulphonylurea-sensitive K(ATP) channels are also present in various intracellular sites including secretory granules, mitochondria, endoplasmic reticulum and more recently, the nucleus. What roles do they play in these organelles? Intracellular K(ATP) channels and sulphonylurea receptors, which operate in conjunction with classical pathways, can provide specific signaling circuits to establish direct links between extracellular signals and different cell functions, such as secretion or gene expression. The study of these intracellular channels provides novel perspectives in the signal transduction of the pancreatic beta-cell, and may offer clues for the development of new strategies in diabetes therapy. In this review we will address this topic with special emphasis on the biophysical basis and functional implications in the pancreatic beta-cell.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas de la Membrana/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Canales de Potasio/metabolismo , Receptores de Droga/metabolismo , Administración Oral , Animales , Sistemas de Liberación de Medicamentos/métodos , Humanos , Líquido Intracelular/efectos de los fármacos , Líquido Intracelular/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Receptores de Sulfonilureas
17.
Diabetologia ; 47(8): 1442-51, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15309294

RESUMEN

AIMS/HYPOTHESIS: We recently demonstrated that insulin-producing cells derived from embryonic stem cells normalise hyperglycaemia in transplanted diabetic mice. The differentiation and selection procedure, however, was successful in less than 5% of the assays performed. Thus, to improve its effectiveness, new strategies have been developed, which increase the number of islet cells or islet progenitors. METHODS: Mouse embryonic stem cells transfected with a plasmid containing the Nkx6.1 promoter gene followed by a neomycin-resistance gene, were cultured with factors known to participate in endocrine pancreatic development and factors that modulate signalling pathways involved in these processes. Neomycin was used to select the Nkx6.1-positive cells, which also express insulin. The transfected cells were differentiated using several exogenous agents, followed by selection of Nkx6.1-positive cells. The resulting cells were analysed for pancreatic gene and protein expression by immunocytochemistry, RT-PCR and radioimmunoassay. Also, proliferation assays were performed, as well as transplantation to streptozotocin-induced diabetic mice. RESULTS: The protocols yielded cell cultures with approximately 20% of cells co-expressing insulin and Pdx-1. Cell trapping selection yielded an almost pure population of insulin-positive cells, which expressed the beta cell genes/proteins Pdx-1, Nkx6.1, insulin, glucokinase, GLUT-2 and Sur-1. Subsequent transplantation to streptozotocin-induced diabetic mice normalised their glycaemia during the time period of experimentation, proving the efficiency of the protocols. CONCLUSIONS/INTERPRETATION: These methods were both highly efficient and very reproducible, resulting in a new strategy to obtain insulin-containing cells from stem cells with a near 100% success rate, while actively promoting the maturation of the exocytotic machinery.


Asunto(s)
Diferenciación Celular/fisiología , Insulina/metabolismo , Células Madre/citología , Animales , Células Cultivadas , Embrión de Mamíferos , Proteínas de Homeodominio/genética , Secreción de Insulina , Islotes Pancreáticos/embriología , Islotes Pancreáticos/metabolismo , Ratones , Regiones Promotoras Genéticas , Transfección
19.
Transplant Proc ; 35(5): 2021-3, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12962883

RESUMEN

In contrast to the consistent observation that methods that promote proliferation also dedifferentiate insulin-producing cells, useful in vitro differentiation protocols must drive both proliferation and differentiation. We herein describe an strategy in which the combination of nutrient restriction and nicotinamide supplementation results in a consistent increase in the mass of insulin-producing cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Insulina/metabolismo , Niacinamida/farmacología , Células Madre/efectos de los fármacos , Células Cultivadas , Humanos , Secreción de Insulina , Células Madre/citología , Células Madre/metabolismo
20.
Med Biol Eng Comput ; 41(4): 384-91, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12892359

RESUMEN

According to the Edmonton protocol, human islet transplantation can result in insulin independency for periods longer than 3 years. However, this therapy for type 1 diabetes is limited by the scarcity of cadaveric donors. Owing to the ability of embryonic stem cells to expand in vitro and differentiate into a variety of cell types, research has focused on ways to manipulate these cells to overcome this problem. It has been demonstrated that mouse embryonic stem cells can differentiate into insulin-containing cells, restoring normoglycaemia in diabetic mice. To this end, mouse embryonic stem cells were transfected with a DNA construct that provides resistance to neomycin under the control of the regulatory regions of the human insulin gene. However, this protocol has a very low efficiency, needing improvements for this technology to be transferred to human stem cells. Optimum protocols will be instrumental in the production of an unlimited source of cells that synthesise, store and release insulin in a physiological manner. The review focuses on the alternative source of tissue offered by embryonic stem cells for regenerative medicine in diabetes and some key points that should be considered in order for a definitive protocol for in vitro differentiation to be established.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Células Madre , Ingeniería de Tejidos/métodos , Técnicas de Cultivo de Célula , Diferenciación Celular , Humanos , Secreción de Insulina , Trasplante de Islotes Pancreáticos , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...