Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 23(4): 2890-2900, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30710421

RESUMEN

Both type 2 diabetes (T2D) and obesity are characterized by excessive hyperlipidaemia and subsequent lipid droplet (LD) accumulation in adipose tissue. To investigate whether LDs also accumulate in ß-cells of T2D patients, we assessed the expression of PLIN2, a LD-associated protein, in non-diabetic (ND) and T2D pancreata. We observed an up-regulation of PLIN2 mRNA and protein in ß-cells of T2D patients, along with significant changes in the expression of lipid metabolism, apoptosis and oxidative stress genes. The increased LD buildup in T2D ß-cells was accompanied by inhibition of nuclear translocation of TFEB, a master regulator of autophagy and by down-regulation of lysosomal biomarker LAMP2. To investigate whether LD accumulation and autophagy were influenced by diabetic conditions, we used rat INS-1 cells to model the effects of hyperglycaemia and hyperlipidaemia on autophagy and metabolic gene expression. Consistent with human tissue, both LD formation and PLIN2 expression were enhanced in INS-1 cells under hyperglycaemia, whereas TFEB activation and autophagy gene expression were significantly reduced. Collectively, these results suggest that lipid clearance and overall homeostasis is markedly disrupted in ß-cells under hyperglycaemic conditions and interventions ameliorating lipid clearance could be beneficial in reducing functional impairments in islets caused by glucolipotoxicity.


Asunto(s)
Autofagia , Diabetes Mellitus Tipo 2/fisiopatología , Regulación de la Expresión Génica , Hiperglucemia/patología , Hiperlipidemias/patología , Células Secretoras de Insulina/patología , Lípidos/análisis , Animales , Apoptosis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hiperglucemia/etiología , Hiperglucemia/metabolismo , Hiperlipidemias/etiología , Hiperlipidemias/metabolismo , Células Secretoras de Insulina/metabolismo , Insulinoma/genética , Insulinoma/metabolismo , Insulinoma/patología , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/metabolismo , Lisosomas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Perilipina-2/genética , Perilipina-2/metabolismo , Ratas , Células Tumorales Cultivadas
2.
Differentiation ; 90(4-5): 77-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26558987

RESUMEN

Regeneration of ß-cells in diabetic patients is an important goal of diabetes research. Islet Neogenesis Associated Protein (INGAP) was discovered in the partially duct-obstructed hamster pancreas. Its bioactive fragment, pentadecapeptide 104-118 (INGAP-P), has been shown to reverse diabetes in animal models and to improve glucose homeostasis in patients with diabetes in clinical trials. Further development of INGAP as a therapy for diabetes requires identification of target cells in the pancreas and characterization of the mechanisms of action. We hypothesized that adult human pancreatic ductal cells retain morphogenetic plasticity and can be induced by INGAP to undergo endocrine differentiation. To test this hypothesis, we treated the normal human pancreatic ductal cell line (HPDE) with either INGAP-P or full-length recombinant protein (rINGAP) for short-term periods. Our data show that this single drug treatment induces both proliferation and transdifferentiation of HPDE cells, the latter being characterized by the rapid sequential activation of endocrine developmental transcription factors Pdx-1, Ngn3, NeuroD, IA-1, and MafA and subsequently the expression of insulin at both the mRNA and the protein levels. After 7 days, C-peptide was detected in the supernatant of INGAP-treated cells, reflecting their ability to secrete insulin. The magnitude of differentiation was enhanced by embedding the cells in Matrigel, which led to islet-like cluster formation. The islet-like clusters cells stained positive for nuclear Pdx-1 and Glut 2 proteins, and were expressing Insulin mRNA. These new data suggest that human adult pancreatic ductal cells retain morphogenetic plasticity and demonstrate that a short exposure to INGAP triggers their differentiation into insulin-expressing cells in vitro. In the context of the urgent search for a regenerative and/or cellular therapy for diabetes, these results make INGAP a promising therapeutic candidate.


Asunto(s)
Antígenos de Neoplasias/farmacología , Biomarcadores de Tumor/farmacología , Péptido C/metabolismo , Proliferación Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Citocinas/farmacología , Diabetes Mellitus/terapia , Insulina/metabolismo , Fragmentos de Péptidos/farmacología , Factores de Transcripción/fisiología , Adulto , Animales , Antígenos de Neoplasias/genética , Biomarcadores de Tumor/genética , Línea Celular , Colágeno/farmacología , Cricetinae , Combinación de Medicamentos , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Laminina/farmacología , Lectinas Tipo C/genética , Conductos Pancreáticos , Proteínas Asociadas a Pancreatitis , Proteoglicanos/farmacología , Proteínas Recombinantes/farmacología
3.
Psychol Trauma ; 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38934934

RESUMEN

OBJECTIVE: Demand for trauma-focused therapy continues to increase, especially in community mental health care settings where group treatment models can be cost-effective and increase access to care. The Internal Family Systems (IFS) model for posttraumatic stress disorder (PTSD) may offer an effective therapeutic approach. The purpose of this proof-of-concept study was to evaluate the feasibility and acceptability of a novel, trauma-focused, group-based treatment approach and investigate potential mechanisms of action. METHOD: Study participants completed the Program for Alleviating and Resolving Trauma and Stress (PARTS), an online-delivered program including 16 weeks of 90-min IFS-based groups with eight 50-min individual IFS counseling sessions. Participants completed assessments including clinician-administered and self-report measures of PTSD, common comorbid conditions (e.g., complex PTSD [disturbances in self-organization], depression, anxiety, and suicidality), and potential mechanisms (e.g., decentering, self-compassion, and emotion regulation). RESULTS: Most participants (n = 11/15; 73%) attended 12+ group sessions, with 92% (12/13 responders) reporting they would recommend PARTS to a friend. All respondents reported the program was helpful (13/13; 100%). PTSD symptom severity was reduced from baseline to Weeks 16 (d = -0.7, p = .005) and 24 (d = -0.9, p < .001). A clinically meaningful response (i.e., 10+ point reduction on the Clinician-Administered PTSD Scale for Diagnostic and Statistical Manual of Mental Disorders [5th ed.]) was demonstrated in 53% of participants (8/15) by Week 24. Decentering, self-compassion, and emotion regulation all improved (p < .05). CONCLUSIONS: PARTS was feasible and acceptable as a group-based, online intervention in an urban, public community health care system. While PARTS showed promise in reducing overall PTSD symptom severity, well-controlled efficacy research is needed. (PsycInfo Database Record (c) 2024 APA, all rights reserved).

4.
Am J Physiol Endocrinol Metab ; 303(7): E917-27, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22850686

RESUMEN

Islet neogenesis-associated protein (INGAP) was discovered in the partially duct-obstructed hamster pancreas as a factor inducing formation of new duct-associated islets. A bioactive portion of INGAP, INGAP(104-118) peptide (INGAP-P), has been shown to have neogenic and insulin-potentiating activity in numerous studies, including recent phase 2 clinical trials that demonstrated improved glucose homeostasis in both type 1 and type 2 diabetic patients. Aiming to improve INGAP-P efficacy and to understand its mechanism of action, we cloned the full-length protein (rINGAP) and compared the signaling events induced by the protein and the peptide in RIN-m5F cells that respond to INGAP with an increase in proliferation. Here, we show that, although both rINGAP and INGAP-P signal via the Ras/Raf/ERK pathway, rINGAP is at least 100 times more efficient on a molar basis than INGAP-P. For either ligand, ERK1/2 activation appears to be pertussis toxin sensitive, suggesting involvement of a G protein-coupled receptor(s). However, there are clear differences between the peptide and the protein in interactions with the cell surface and in the downstream signaling. We demonstrate that fluorescent-labeled rINGAP is characterized by clustering on the membrane and by slow internalization (≤5 h), whereas INGAP-P does not cluster and is internalized within minutes. Signaling by rINGAP appears to involve Src, in contrast to INGAP-P, which appears to activate Akt in addition to the Ras/Raf/ERK1/2 pathway. Thus our data suggest that interactions of INGAP with the cell surface are important to consider for further development of INGAP as a pharmacotherapy for diabetes.


Asunto(s)
Antígenos de Neoplasias/farmacología , Biomarcadores de Tumor/farmacología , Citocinas/farmacología , Islotes Pancreáticos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Clonación Molecular , Lectinas Tipo C , Proteínas Asociadas a Pancreatitis , Toxina del Pertussis/farmacología , Ratas , Transducción de Señal/efectos de los fármacos
5.
Biotechnol Appl Biochem ; 59(2): 106-20, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23586791

RESUMEN

Human mesenchymal stem cells (hMSCs) have many potential applications in tissue engineering and regenerative medicine. Currently, hMSCs are generated through conventional static adherent cultures in the presence of fetal bovine serum (FBS) for clinical applications (e.g., multiple sclerosis). However, these methods are not appropriate to meet the expected future demand for quality-assured hMSCs for human therapeutic use. Hence, it is imperative to develop an effective hMSC production system, which should be controllable, reproducible, and scalable. To this end, efforts have been made by several international research groups to develop (i) alternative media either by replacing FBS with human-sourced supplements (such as human serum or platelet lysate) or by identifying defined serum-free formulations consisting of key growth/attachment factors, and (ii) controlled bioreactor protocols. In this regard, we review here current hMSC production technologies and future perspectives toward efficient methods for the generation of clinically relevant numbers of hMSC therapeutics.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/citología , Biotecnología/métodos , Medios de Cultivo , Humanos
6.
Cell Death Discov ; 7(1): 56, 2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731692

RESUMEN

The goal of this study was to determine whether recombinant Islet NeoGenesis Associated Protein (rINGAP) and its active core, a pentadecapeptide INGAP104-118 (Ingap-p), protect ß cells against cytokine-induced death. INGAP has been shown to induce islet neogenesis in diabetic animals, to stimulate ß-cell proliferation and differentiation, and to improve islet survival and function. Importantly, Ingap-p has shown promising results in clinical trials for diabetes (phase I/II). However, the full potential of INGAP and its mechanisms of action remain poorly understood. Using rat insulinoma cells RINm5F and INS-1 treated with interleukin-1ß (IL-1ß) and interferon-gamma (IFN-γ), we demonstrate here that both rINGAP and Ingap-p inhibit apoptosis, Caspase-3 activation, inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production, and explore the related signaling pathways. As expected, IL-1ß induced nuclear factor kappa B (NF-κB), p38, and JNK signaling, whereas interferon-gamma (IFN-γ) activated the JAK2/STAT1 pathway and potentiated the IL-1ß effects. Both rINGAP and Ingap-p decreased phosphorylation of IKKα/ß, IkBα, and p65, although p65 nuclear translocation was not inhibited. rINGAP, used for further analysis, also inhibited STAT3, p38, and JNK activation. Interestingly, all inhibitory effects of rINGAP were observed for the cytokine cocktail, not IL-1ß alone, and were roughly equal to reversing the potentiating effects of INFγ. Furthermore, rINGAP had no effect on IL-1ß/NF-κB-induced gene expression (e.g., Ccl2, Sod2) but downregulated several IFNγ-stimulated (Irf1, Socs1, Socs3) or IFNγ-potentiated (Nos2) genes. This, however, was observed again only for the cytokine cocktail, not IFNγ alone, and rINGAP did not inhibit the IFNγ-induced JAK2/STAT1 activation. Together, these intriguing results suggest that INGAP does not target either IL-1ß or IFNγ individually but rather inhibits the signaling crosstalk between the two, the exact mechanism of which remains to be investigated. In summary, our study characterizes the anti-inflammatory effects of INGAP, both protein and peptide, and suggests a new therapeutic utility for INGAP in the treatment of diabetes.

7.
Biochim Biophys Acta ; 1790(6): 566-74, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19264107

RESUMEN

BACKGROUND: Amyloid fibrils created by misfolding and aggregation of proteins are a major pathological feature in a variety of degenerative diseases. Therapeutic approaches including amyloid vaccines and anti-aggregation compounds in models of amyloidosis point to an important role for amyloid in disease pathogenesis. Amyloid deposits derived from the beta-cell peptide islet amyloid polypeptide (IAPP or amylin) are a characteristic of type 2 diabetes and may contribute to loss of beta-cells in this disease. METHODS: We developed a cellular model of rapid amyloid deposition using cultured human islets and observed a correlation between fibril accumulation and beta-cell death. A series of overlapping peptides derived from IAPP was generated. RESULTS: A potent inhibitor (ANFLVH) of human IAPP aggregation was identified. This inhibitory peptide prevented IAPP fibril formation in vitro and in human islet cultures leading to a striking increase in islet cell viability. CONCLUSIONS: These findings indicate an important contribution of IAPP aggregation to beta-cell death in situ and point to therapeutic applications for inhibitors of IAPP aggregation in enhancing beta-cell survival. GENERAL SIGNIFICANCE: Anti-amyloid compounds could potentially reduce the loss of beta-cell mass in type 2 diabetes and maintain healthy human islet cultures for beta-cell replacement therapies.


Asunto(s)
Amiloide/antagonistas & inhibidores , Supervivencia Celular/fisiología , Islotes Pancreáticos , Secuencia de Aminoácidos , Amiloide/metabolismo , Animales , Apoptosis/fisiología , Células Cultivadas , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos , Islotes Pancreáticos/fisiología , Islotes Pancreáticos/ultraestructura , Datos de Secuencia Molecular
8.
J Cell Physiol ; 224(2): 501-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20432450

RESUMEN

Type 1 diabetes is inhibited in diabetes-prone BioBreeding (BBdp) rats fed a low-antigen hydrolyzed casein (HC) diet. In cereal-fed BBdp rats, islet expansion is defective accompanied by a futile upregulation of islet neogenesis without increased islet mass, due to a subtle blockage in islet cell cycle. We hypothesized that islet growth is enhanced before insulitis in HC-fed young BBdp rats and that islet neogenesis could be stimulated by a trophic factor, islet neogenesis-associated protein (INGAP). beta-Cell homeostasis was analyzed using immunohistochemistry, morphometry, laser capture microdissection and RT-PCR in BBdp rats fed HC or cereal diets. beta-cell proliferation in small and medium islets, and the number and area fraction of medium and large islets were increased in HC-fed animals. In situ islet cell cycle analysis revealed an increased proportion of proliferating S + G2 cells in medium and large islets of 25-45 day HC-fed rats. Expression of the cell cycle inhibitor, p16(INK4a) correlated with islet size and the percentage of p16(INK4a+) beta-cells increased in HC-fed BBdp rats, likely reflecting an increase in large islet area fraction. In HC-fed rats, extra-islet insulin(+) clusters (EIC), insulin(+) duct cells, large islet area fraction, and beta-cell mass were increased. Neurogenin-3 and Pdx-1, markers of beta-cell progenitors, were increased in EIC of weanling HC-fed rats. Daily injection of INGAP (30-45 days) increased the number of small islets, total islets, and insulin(+) cells in small ducts. Thus, in BBdp rats fed a protective HC diet, beta-cell expansion is enhanced through increased beta-cell proliferation and stimulation of islet neogenesis.


Asunto(s)
Envejecimiento/patología , Citoprotección , Diabetes Mellitus Experimental/patología , Dieta , Conducta Alimentaria , Células Secretoras de Insulina/patología , Envejecimiento/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Caseínas/administración & dosificación , Caseínas/farmacología , Agregación Celular/efectos de los fármacos , Recuento de Células , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Citocinas/farmacología , Citoprotección/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Susceptibilidad a Enfermedades/patología , Proteínas de Homeodominio/metabolismo , Homeostasis/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Proteínas Asociadas a Pancreatitis , Fragmentos de Péptidos/farmacología , Ratas , Transactivadores/metabolismo
9.
Cytotherapy ; 12(5): 637-57, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20608762

RESUMEN

BACKGROUND AIMS: Ex vivo propagation of sparse populations of human mesenchymal stromal cells (hMSC) is critical for generating numbers sufficient for therapeutic applications. hMSC culture media have typically been supplemented with animal serum and, recently, human-sourced materials. However, these supplements are ill-defined and, thus, undesirable for clinical and research applications. Previously reported efforts to develop defined media for hMSC culture only resulted in slow or limited proliferation, and were unsuccessful in expanding these cells from primary cultures. Therefore a major step forward would be the identification of defined, serum-free culture conditions capable of supporting both the isolation and rapid expansion of hMSC. METHODS: Using classical approaches of medium development, we were able to identify a set of growth and attachment factors that allowed the serum-free isolation and expansion of hMSC from bone marrow. RESULTS: Heparin, selenium and platelet-derived growth factor (PDGF)-BB were found to be inhibitory for the growth of hMSC, whereas basic fibroblast growth factor (bFGF) was critical and worked synergistically with transforming growth factor (TGF)-beta1 to allow significant cell expansion. Ascorbic acid, hydrocortisone and fetuin were also found to be important growth and attachment factors that, in conjunction with substrate-coating proteins, allowed the isolation of hMSC from primary culture and their subsequent expansion. CONCLUSIONS: We report a defined medium formulation (PPRF-msc6), consisting of key recombinant and serum-derived components, for the rapid isolation and expansion of hMSC in the absence of serum. This work represents an important step forward for achieving an ideal, completely defined synthetic medium composition for the safe use of hMSC in clinical settings.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Células del Estroma/metabolismo , Ácido Ascórbico/farmacología , Médula Ósea/patología , Adhesión Celular/efectos de los fármacos , Separación Celular , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/farmacología , Citometría de Flujo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Factor de Crecimiento Transformador beta/farmacología , alfa-Fetoproteínas/farmacología
10.
Protein Expr Purif ; 69(1): 1-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19635567

RESUMEN

Islet Neogenesis Associated Protein (INGAP) is implicated in pancreatic islet neogenesis. INGAP peptide, a pentadecapeptide comprising amino acids 104-118, reverses diabetes in rodents and improves glucose homeostasis in patients with diabetes. The mechanism of INGAP action is unknown, but such studies would benefit from the availability of the full-length recombinant protein (rINGAP). Here we report the production of rINGAP from 293-SF cells following lentiviral transduction, and its characterization by MALDI-TOF and Q-TOF Mass Spectrometry, and HPLC. Importantly, we show that rINGAP exhibits 100x the bioactivity of INGAP peptide on a molar basis in an in vitro assay of human islet regeneration.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Biomarcadores de Tumor/biosíntesis , Lectinas Tipo C/biosíntesis , Proteínas Recombinantes/biosíntesis , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/aislamiento & purificación , Biomarcadores de Tumor/química , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/aislamiento & purificación , Células Cultivadas , Cromatografía Líquida de Alta Presión , Cricetinae , Regulación de la Expresión Génica , Humanos , Islotes Pancreáticos/fisiología , Lectinas Tipo C/química , Lectinas Tipo C/genética , Lectinas Tipo C/aislamiento & purificación , Lentivirus/genética , Espectrometría de Masas , Mesocricetus , Datos de Secuencia Molecular , Peso Molecular , Proteínas Asociadas a Pancreatitis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Regeneración/fisiología , Fracciones Subcelulares/metabolismo , Transducción Genética
11.
Methods Mol Biol ; 482: 371-85, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19089368

RESUMEN

Current therapies do not prevent the complications of diabetes. Furthermore, these therapies do not address the underlying pathology; the lack of functional beta-cell mass that occurs in both types 1 and 2 diabetes. While pancreas and islet transplantation do serve to increase beta-cell mass, a lack of donor organs limits the therapeutic potential of these treatments. As such, expansion of beta-cell mass from endogenous sources, either in vivo or in vitro, represents an area of increasing interest. One potential source of islet progenitors is the islet proper, via the dedifferentiation, proliferation, and redifferentiation of facultative progenitors residing within the islet. We have developed a tissue culture platform whereby isolated adult human pancreatic islets form proliferative duct-like structures expressing ductal and progenitor markers. Short-term treatment with a peptide fragment of islet neogenesis-associated protein (INGAP) induces these structures to reform islet-like structures that resemble freshly isolated islets with respect to the frequency and distribution of the four endocrine cell types, islet gene expression and hormone production, insulin content, and glucose-responsive insulin secretion. As such, the plasticity of adult human islets has significant implications for islet regeneration.


Asunto(s)
Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Regeneración , Células Madre/citología , Ingeniería de Tejidos/métodos , Animales , Células Cultivadas , Humanos , Inmunohistoquímica , Proteínas Asociadas a Pancreatitis , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Adhesión del Tejido
12.
Biochim Biophys Acta ; 1769(9-10): 579-85, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17673309

RESUMEN

The regenerating (Reg) genes are associated with tissue repair and have been directly implicated in pancreatic beta-cell regeneration. A hamster Reg3, Islet neogenesis associated protein (INGAP), has been shown to possess anti-diabetic properties in rodent models. Although several Reg3 proteins have been identified in other species, INGAP is the only Reg3 found in hamsters. To identify new Reg3 genes in the hamster pancreas we employed homology reverse transcription polymerase chain reaction (RT-PCR) using degenerate Reg3 primers, followed by rapid amplification of cDNA ends (RACE). We report here the discovery of a new hamster Reg3 gene of 765 nucleotides (nt) that encodes a 174-amino acid (aa) protein. This protein sequence was identified as a novel hamster Reg3gamma with 78% and 75% identity to the rat Reg3gamma and mouse Reg3gamma protein, respectively. We also fully sequenced the previously reported partial sequence of the hamster Reg1 gene coding region using RACE to yield a 756-nt transcript that encodes a deduced 173 aa protein. This protein was identified as hamster Reg2, rather than Reg1 as was initially reported, with an 81% identity to mouse Reg2. The spatial gene expression patterns of the hamster Reg genes, analyzed by RT-PCR, were similarly distributed with low level expression being found globally throughout the body. Mice and hamsters are the only species known to carry either of the functional INGAP or Reg2 genes. It remains to be determined whether these genes bestow mice and hamsters with special regenerative abilities in the pancreas.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Células Secretoras de Insulina/fisiología , Proteínas/genética , Regeneración/fisiología , Animales , Secuencia de Bases , Clonación Molecular , Cricetinae , ADN Complementario/genética , Mesocricetus , Ratones , Datos de Secuencia Molecular , Especificidad de Órganos/fisiología , Proteínas Asociadas a Pancreatitis , Proteínas/metabolismo , Homología de Secuencia de Ácido Nucleico
13.
Lab Invest ; 88(7): 761-72, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18490899

RESUMEN

Cultured human islets can be dedifferentiated to duct-like structures composed mainly of cytokeratin+ and nestin+ cells. Given that these structures possess the potential to redifferentiate into islet-like structures, we sought to elucidate their specific cellular origins. Adenoviral vectors were engineered for beta-, alpha-, delta- or PP-cell-specific GFP expression. A double-stranded system was designed whereby cultures were infected with two vectors: one expressed GFP behind the cumate-inducible promoter sequence, and the other expressed the requisite transactivator behind the human insulin, glucagon, somatostatin or pancreatic polypeptide promoter. This system labels hormone+ cells in the islet in a cell-specific manner, allowing these cells to be tracked during the course of transformation from islet to duct-like structure. Post-infection, islets were cultured to induce dedifferentiation. Fluorescence microscopy demonstrated that alpha-, delta- and PP-cells contributed equally to the cytokeratin+ population, with minimal beta-cell contribution, whereas the converse was true for nestin+ cells. Complementary targeted cell ablation studies, using streptozotocin or similar adenoviral expression of the Bax (Bcl2-associated X protein) toxigene, validated these findings and suggested a redundancy between alpha-, delta- and PP-cells with respect to cytokeratin+ cell derivation. These results call into question the traditional understanding of islet cells as being terminally differentiated and provide support for the concept of adult islet morphogenetic plasticity.


Asunto(s)
Islotes Pancreáticos/citología , Adenoviridae/genética , Adulto , Diferenciación Celular , Células Cultivadas , Vectores Genéticos , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Islotes Pancreáticos/metabolismo , Queratinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nestina , Conductos Pancreáticos/citología , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/metabolismo , Regiones Promotoras Genéticas , Somatostatina/fisiología , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/metabolismo , Células Madre/citología , Estreptozocina/farmacología , Proteína X Asociada a bcl-2/biosíntesis , Proteína X Asociada a bcl-2/genética
14.
Transplantation ; 85(7): 950-5, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18408573

RESUMEN

BACKGROUND: Recent advances in the fields of islet transplantation and in vitro islet cell expansion place a renewed emphasis on the optimization of islet isolation from cadaveric human donor organs. We retrospectively analyzed 171 islet isolations to identify variables that predict islet yield and isolation success. METHODS: Cadaveric human donor pancreata were procured and processed according to established protocols. Donor-, procurement-, and isolation-related variables were analyzed for correlation with islet yield and isolation success (> or =250,000 islet equivalents). RESULTS: Univariate analysis suggested correlations between islet yield and donor age (P<0.005), body surface area (P<0.005), duration of enzymatic digestion (P<0.001), and pancreatic beta-cell volume (P<0.05). Donor sex (P<0.01), procurement team (P<0.05), and peridigestion serine protease inhibition (P<0.05) affected islet yield, whereas enzyme lot (P<0.01) and pancreatic fatty infiltration (P<0.05) influenced isolation success. By logistic regression, donor sex and age, and duration of enzymatic digestion could predict a successful isolation with 72% accuracy. The use of Liberase CI improved islet yield (P<0.05) in young donors (< or =25 years). CONCLUSIONS: While donor-related variables are useful in predicting islet yield, these are likely surrogates for pancreatic beta-cell volume. Enzyme lot, and the associated duration of enzymatic digestion (P<0.05), appears to be key determinants of isolation success.


Asunto(s)
Islotes Pancreáticos/citología , Donantes de Tejidos , Adulto , Índice de Masa Corporal , Cadáver , Separación Celular , Femenino , Humanos , Células Secretoras de Insulina/citología , Masculino , Persona de Mediana Edad , Selección de Paciente , Análisis de Regresión , Recolección de Tejidos y Órganos/métodos
15.
Mol Endocrinol ; 21(6): 1467-77, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17405902

RESUMEN

Tissue plasticity is well documented in the context of pancreatic regeneration and carcinogenesis, with recent reports implicating dedifferentiated islet cells both as endocrine progenitors and as the cell(s) of origin in pancreatic adenocarcinoma. Accordingly, it is noteworthy that accumulating evidence suggests that TGFbeta signaling is essential to pancreatic endocrine development and maintenance, whereas its loss is associated with the progression to pancreatic adenocarcinoma. The aim of this study was to examine the role of TGFbeta in an in vitro model of islet morphogenetic plasticity. Human islets were embedded in a collagen gel and cultured under conditions that induced transformation into duct-like epithelial structures (DLS). Addition of TGFbeta caused a dose-dependent decrease in DLS formation. Although it was demonstrated that collagen-embedded islets secrete low levels of TGFbeta, antibody-mediated neutralization of this endogenously released TGFbeta improved DLS formation rates, suggesting local TGFbeta concentrations may in fact be higher. Time course studies indicated that TGFbeta signaling was associated with an increase in ERK and p38 MAPK phosphorylation, although inhibitor-based studies were consistent with an islet endocrine-stabilizing effect mediated by p38 alone. Localization of TGFbeta signaling molecules suggested that the action of TGFbeta is directly on the beta-cell to inhibit apoptosis and thus stabilize endocrine phenotype.


Asunto(s)
Islotes Pancreáticos/fisiología , Factor de Crecimiento Transformador beta/fisiología , Adulto , Diferenciación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Islotes Pancreáticos/química , Islotes Pancreáticos/efectos de los fármacos , MAP Quinasa Quinasa 4/metabolismo , Conductos Pancreáticos/citología , Conductos Pancreáticos/crecimiento & desarrollo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Healthc Pap ; 17(4): 32-40, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-30291708

RESUMEN

In hindsight, there have been unintended systemic consequences stemming from the traditional roles physicians have assumed and the structures within which they have been permitted to organize themselves. It is critical that the national discussion take account of this because we must reconcile ourselves to the current reality in which all other allied healthcare professionals are practising at "the top of their licence." Furthermore, the pace of technological change, especially the deciphering of the genome and the digitalization of virtually everything, has engendered a revolution characterized by the democratization of knowledge and technology, so that the point of care will be wherever the patient is. Dysfunctional reimbursement schemes and a lack of accountability are merely symptoms of a system that must change.


Asunto(s)
Atención a la Salud/métodos , Reforma de la Atención de Salud , Médicos , Responsabilidad Social , Humanos , Quebec
17.
Cell Biochem Biophys ; 48(2-3): 127-37, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17709882

RESUMEN

Islet Neogenesis-Associated Protein (INGAP) is a member of the Reg family of proteins implicated in various settings of endogenous pancreatic regeneration. The expression of INGAP and other RegIII proteins has also been linked temporally and spatially with the induction of islet neogenesis in animal models of disease and regeneration. Furthermore, administration of a peptide fragment of INGAP (INGAP peptide) has been demonstrated to reverse chemically induced diabetes as well as improve glycemic control and survival in an animal model of type 1 diabetes. Cultured human pancreatic tissue has also been shown to be responsive to INGAP peptide, producing islet-like structures with function, architecture and gene expression matching that of freshly isolated islets. Likewise, studies in normoglycemic animals show evidence of islet neogenesis. Finally, recent clinical studies suggest an effect of INGAP peptide to improve insulin production in type 1 diabetes and glycemic control in type 2 diabetes.


Asunto(s)
Antígenos de Neoplasias/fisiología , Biomarcadores de Tumor/fisiología , Islotes Pancreáticos/fisiología , Lectinas Tipo C/fisiología , Regeneración/fisiología , Animales , Antígenos de Neoplasias/farmacología , Biomarcadores de Tumor/farmacología , Ensayos Clínicos Fase II como Asunto , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Páncreas/fisiología , Proteínas Asociadas a Pancreatitis , Regeneración/efectos de los fármacos
19.
Int J Biochem Cell Biol ; 38(4): 498-503, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16216541

RESUMEN

Current therapies for type 1 diabetes, including fastidious blood glucose monitoring and multiple daily insulin injections, are not sufficient to prevent complications of the disease. Though pancreas and possibly islet transplantation can prevent the progression of complications, the scarcity of donor organs limits widespread application of these approaches. Understanding the mechanisms of beta-cell mass expansion as well as the means to exploit these pathways has enabled researchers to develop new strategies to expand and maintain islet cell mass. Potential new therapeutic avenues include ex vivo islet expansion and improved viability of islets prior to implantation, as well as the endogenous expansion of beta-cell mass within the diabetic patient. Islet neogenesis, through stem cell activation and/or transdifferentiation of mature fully differentiated cells, has been proposed as a means of beta-cell mass expansion. Finally, any successful new therapy for type 1 diabetes via beta-cell mass expansion will require prevention of beta-cell death and maintenance of long-term endocrine function.


Asunto(s)
Proliferación Celular , Complicaciones de la Diabetes/terapia , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/trasplante , Recuperación de la Función , Donantes de Tejidos , Animales , Automonitorización de la Glucosa Sanguínea , Técnicas de Cultivo de Célula , Muerte Celular , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Complicaciones de la Diabetes/patología , Diabetes Mellitus Tipo 1/patología , Humanos , Insulina/administración & dosificación , Células Secretoras de Insulina/patología , Trasplante de Islotes Pancreáticos , Ratones , Factores de Tiempo , Donantes de Tejidos/provisión & distribución
20.
Transplantation ; 82(6): 813-8, 2006 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-17006329

RESUMEN

BACKGROUND: Recent successes in islet transplantation highlight the importance of islet isolation by experienced centers and minimization of cell injury as crucial to the achievement of insulin independence. Islet injury may manifest as cell death by apoptosis, shorter graft survival, and the need for retransplantation. Although an inflammatory cytokine response at the graft site is known to inhibit engraftment, recent evidence indicates that islet cells may contribute to this response. METHODS: Isolated human islets were cultured for up to one week in serum-free CMRL-1066 with 25 microM of tumor necrosis factor (TNF)alpha inhibitor RDP58. Gene expression was measured by reverse transcriptase polymerase chain reaction, apoptosis and TNFalpha secretion by enzyme-linked immunosorbent assay and enzyme-linked immunospot, and islet function by stimulated insulin secretion. RESULTS: Isolation induced a twofold increase in TNFalpha expression between days one and three (P<0.05), while TNFalpha secretion peaked at day one. RDP58 reduced TNFalpha secretion by 70.6% (P<0.02), though TNFalpha gene expression was unaffected. RDP58 reduced the frequency of TNFalpha-secreting islets by 64.4% (P<0.05) and reduced apoptotic levels by 26.4% within 24 hr postisolation (P<0.05). The reduction in apoptosis was maintained throughout the week (P<0.01), while apoptosis increased in control cultures. Finally, RDP58-treated islets displayed increased insulin secretion in response to both elevated glucose (1915.0+/-396.6 vs. 825.3+/-261.1 mU/L, P<0.01) and secretagogues (2294.3+/-529.5 vs. 939.8+/-333.7 mU/L, P<0.02). CONCLUSIONS: These data demonstrate that intraislet cytokine production should be considered as a factor leading to islet cell death postisolation and postengraftment, and strategies aimed at countering islet cytokine production represent a novel target for improving islet viability and function.


Asunto(s)
Muerte Celular/fisiología , Islotes Pancreáticos/fisiología , Factor de Necrosis Tumoral alfa/genética , Adulto , Apoptosis , Cadáver , Células Cultivadas , Cartilla de ADN , Femenino , Regulación de la Expresión Génica , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Masculino , Persona de Mediana Edad , Péptidos/farmacología , Donantes de Tejidos , Transcripción Genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA