Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell Mol Neurobiol ; 40(3): 369-380, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31522299

RESUMO

Besides other causes, ischemia and Alzheimer's disease pathology is also linked to decreased cerebral blood flow (CBF). There is little or no consensus about the role of neuroglial cells in maintaining CBF in various neuropathologies. This consensus becomes scarcer when it comes to clinical and experimental cases of comorbid Abeta-amyloid (Aß) toxicity and ischemia. Here, a comorbid rat model of Aß toxicity and endothelin-1 induced ischemia (ET1) not only demonstrated the appearance of axotomized phagocytosed pyknotic neurons (NeuN) immediately after the injury, but also showed a diversity of continuously changing neuroglia (MHC Class II/OX6, Iba1) and macrophage (Iba1/CD68) phenotypes with round, stout somas, and retracted processes. This is indicative of a response to a concomitant increase in large fluid-filled spaces due to the vascular leakage. Ironically 4 weeks after the injury despite a conclusive reduction in neurons, CBF restoration in ET1 rats was associated with a massive increase in neuroglial cell numbers, hypertrophy, ramification, and soma sizes bordering the continuously reducing lesion core and inflamed vasculature, possibly to shield their leaky phenotype. Astrocytes were also found to be releasing matrix metalloproteinase9 (MMP9), which stabilized matrix ligand ß-dystroglycan (ß-DG) in repaired or functional vessels. Changing neuroglia phenotypes, responses, motility, astrocytic recruitment of MMP9, and ß-DG stabilization implies the role of communication between neuroglia and endothelium in recovering CBF, in the absence of neurons, in ET1 rats compared to Aß+ET1 rats, which showed characteristics delayed neuroglial activation. Stimulation of timely neuroglial reactivity may serve as a viable strategy to compensate for the neuronal loss in restoring CBF in comorbid cases of ischemia and Aß toxicity.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Circulação Cerebrovascular/fisiologia , Neuroglia/fisiologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides , Animais , Astrócitos/patologia , Astrócitos/fisiologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Encéfalo/fisiopatologia , Isquemia Encefálica/fisiopatologia , Modelos Animais de Doenças , Masculino , Neurônios/patologia , Neurônios/fisiologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia
2.
Cell Mol Neurobiol ; 40(3): 381-382, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31599387

RESUMO

The original version of this article contained a random order of part labels for Fig. 4. The correct caption of Fig. 4 with correct order of part labels is given below.

3.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30217817

RESUMO

Fibroblast growth factor receptor-1 (FGFR1) activity at the plasma membrane is tightly controlled by the availability of co-receptors and competing receptor isoforms. We have previously shown that FGFR1 activity in pancreatic beta-cells modulates a wide range of processes, including lipid metabolism, insulin processing, and cell survival. More recently, we have revealed that co-expression of FGFR5, a receptor isoform that lacks a tyrosine-kinase domain, influences FGFR1 responses. We therefore hypothesized that FGFR5 is a co-receptor to FGFR1 that modulates responses to ligands by forming a receptor heterocomplex with FGFR1. We first show here increased FGFR5 expression in the pancreatic islets of nonobese diabetic (NOD) mice and also in mouse and human islets treated with proinflammatory cytokines. Using siRNA knockdown, we further report that FGFR5 and FGFR1 expression improves beta-cell survival. Co-immunoprecipitation and quantitative live-cell imaging to measure the molecular interaction between FGFR5 and FGFR1 revealed that FGFR5 forms a mixture of ligand-independent homodimers (∼25%) and homotrimers (∼75%) at the plasma membrane. Interestingly, co-expressed FGFR5 and FGFR1 formed heterocomplexes with a 2:1 ratio and subsequently responded to FGF2 by forming FGFR5/FGFR1 signaling complexes with a 4:2 ratio. Taken together, our findings identify FGFR5 as a co-receptor that is up-regulated by inflammation and promotes FGFR1-induced survival, insights that reveal a potential target for intervention during beta-cell pathogenesis.


Assuntos
Citocinas/imunologia , Diabetes Mellitus/genética , Células Secretoras de Insulina/imunologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Animais , Diabetes Mellitus/imunologia , Dimerização , Feminino , Fator 2 de Crescimento de Fibroblastos/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/imunologia , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/imunologia , Regulação para Cima
4.
J Endocrinol ; 260(1)2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37855335

RESUMO

Reports in North America suggest that up to 20% of young women (18-24 years) use cannabis during pregnancy. This is concerning given clinical studies indicate that maternal cannabis use is associated with fetal growth restriction and dysglycemia in the offspring. Preclinical studies demonstrated that prenatal exposure to Δ9-tetrahydrocannabinol, the main psychoactive component of cannabis, in rat dams led to female-specific deficits in ß-cell mass and glucose intolerance/insulin resistance. Yet to date, the contributions of cannabidiol (CBD), the primary nonpsychoactive compound in cannabis, remain elusive. This study aimed to define the effects of in utero cannabidiol (CBD) exposure on postnatal glucose regulation. Pregnant Wistar rat dams received daily intraperitoneal injections of either a vehicle solution or 3 mg/kg of CBD from gestational day (GD) 6 to parturition. CBD exposure did not lead to observable changes in maternal or neonatal outcomes; however, by 3 months of age male CBD-exposed offspring exhibited glucose intolerance despite no changes in pancreatic ß/α-cell mass. Transcriptomic analysis on the livers of these CBD-exposed males revealed altered gene expression of circadian rhythm clock machinery, which is linked to systemic glucose intolerance. Furthermore, alterations in hepatic developmental and metabolic processes were also observed, suggesting gestational CBD exposure has a long-lasting detrimental effect on liver health throughout life. Collectively, these results indicate that exposure to CBD alone in pregnancy may be detrimental to the metabolic health of the offspring later in life.


Assuntos
Canabidiol , Intolerância à Glucose , Resistência à Insulina , Células Secretoras de Insulina , Gravidez , Ratos , Feminino , Masculino , Humanos , Animais , Lactente , Canabidiol/toxicidade , Intolerância à Glucose/induzido quimicamente , Ratos Wistar
5.
J Comp Neurol ; 529(2): 421-433, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32447764

RESUMO

Combination of ischemia and ß-amyloid (Aß) toxicity has been shown to simultaneously increase neuro-inflammation, endogenous Aß deposition, and neurodegeneration. However, studies on the evolution of infarct and panorama of cellular degeneration as a synergistic or overlapping mechanism between ischemia and Aß toxicity are lacking. Here, we compared fluorojade B (FJB) and hematoxylin and eosin (H&E) stains primarily to examine the chronology of infarct, and the viability and morphological changes in neuroglia and neurons located in different brain regions on d1, d7, and d28 post Aß toxicity and endothelin-1 induced ischemia (ET1) in rats. We demonstrated a regional difference in cellular degeneration between cortex, corpus callosum, striatum, globus pallidus, and thalamus after cerebral injury. Glial cells in the cortex and corpus callosum underwent delayed FJB staining from d7 to d28, but neurons in cortex disappeared within the first week of cerebral injury. Striatal lesion core and globus pallidus of Aß + ET1 rats showed extensive degeneration of neuronal cells compared with ET1 rats alone starting from d1. Differential and exacerbated expressions of cyclooxygenase-2 might be the cause of excessive neuronal demise in the striatum of Aß + ET1 rats. Such an investigation may improve our understanding to identify and manipulate a critical therapeutic window post comorbid injury.


Assuntos
Lesões Encefálicas/patologia , Sobrevivência Celular/fisiologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiologia , Degeneração Neural/patologia , Animais , Córtex Cerebral/química , Corpo Estriado/química , Corpo Estriado/patologia , Corpo Estriado/fisiologia , Masculino , Ratos , Ratos Wistar
6.
Pediatr Res ; 68(1): 16-22, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20386490

RESUMO

Beta cells are partially replaced in neonatal rodents after deletion with streptozotocin (STZ). Exposure of pregnant rats to a low protein (LP) diet impairs endocrine pancreas development in the offspring, leading to glucose intolerance in adulthood. Our objective was to determine whether protein restriction has a similar effect on the offspring in mice, and if this alters the capacity for beta cell regeneration after STZ. Pregnant Balb/c mice were fed a control (C) (20% protein) or an isocaloric LP (8% protein) diet during gestation. Pups were given 35 mg/kg STZ (or vehicle) from d 1 to 5 for each dietary treatment. Histologic analysis showed that C-fed offspring had largely replaced beta cell mass (BCM) after STZ by d 30, but this was not sustained over time. Female LP-fed offspring showed an initial increase in BCM by d 14 but developed glucose intolerance by d 130. In contrast, male LP offspring showed no changes in BCM or glucose tolerance. However, LP exposure limited the capacity for recovery of BCM in both genders after STZ treatment.


Assuntos
Proteínas Alimentares/metabolismo , Células Secretoras de Insulina/fisiologia , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Regeneração/fisiologia , Animais , Glicemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/metabolismo , Dieta , Feminino , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão , Pâncreas/metabolismo , Gravidez , Distribuição Aleatória , Ratos
7.
J Comp Neurol ; 528(1): 48-60, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31265125

RESUMO

Large cerebral infarctions are major predictors of death and severe disability from stroke. Conversely, data concerning these types of infarctions and the affected adjacent brain circuits are scarce. It remains to be determined if the co-morbid concurrence of large infarct and ß-amyloid (Aß) toxicity can precipitate the early development of dementia. Here, we described a dose-dependent effect of a unilateral striatal injection of vasoconstrictive endothelin-1 (ET-1) along with Aß toxicity on CNS pathogenesis; driven by the anatomical and functional networks within a brain circuit. After 21 days of treatment, a high dose (60 pmol) of ET-1 (E60) alone caused the greatest increase in neuroinflammation, mainly in the ipsilateral striatum and distant regions with synaptic links to the striatal lesion such as white matter (subcortical white matter, corpus callosum, internal capsule, anterior commissure), gray matter (globus pallidus, thalamus), and cortices (cingulate, motor, somatosensory, entorhinal). The combined E60 + Aß treatment also extended perturbation in the contralateral hemisphere of these rats, such as increased deposition of amyloid precursor protein fragments associated with the appearance of degenerating cells and the leakage of laminin from the basement membrane across a compromised blood-brain barrier. However, the cerebral damage induced by the 6 pmol ET-1 (E6), Aß and E6 + Aß rats was not detrimental enough to injure the complete network. The appreciation of the causal interactions among distinct anatomical units in the brain after ischemia and Aß toxicity will help in the design of effective and alternative therapeutics that may disassociate the synergistic or additive association between the infarcts and Aß toxicity.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Encéfalo/patologia , Infarto Cerebral/patologia , Endotelina-1/toxicidade , Rede Nervosa/patologia , Peptídeos beta-Amiloides/administração & dosagem , Animais , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Infarto Cerebral/induzido quimicamente , Endotelina-1/administração & dosagem , Injeções Intraventriculares , Masculino , Rede Nervosa/anatomia & histologia , Rede Nervosa/efeitos dos fármacos , Ratos , Ratos Wistar
8.
Mol Neurobiol ; 56(4): 2714-2727, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30054859

RESUMO

The hippocampus, a brain region vital for memory and learning, is sensitive to the damage caused by ischemic/hypoxic stroke and is one of the main regions affected by Alzheimer's disease. The pathological changes that might occur in the hippocampus and its connections, because of cerebral injury in a distant brain region, such as the striatum, have not been examined. Therefore, in the present study, we evaluated the combined effects of endothelin-1-induced ischemia (ET1) in the striatum and ß-amyloid (Aß) toxicity on hippocampal pathogenesis, dictated by the anatomical and functional intra- and inter-regional hippocampal connections to the striatum. The hippocampal pathogenesis induced by Aß or ET1 alone was not severe enough to significantly affect the entire circuit of the hippocampal network. However, the combination of the two pathological states (ET1 + Aß) led to an exacerbated increase in neuroinflammation, deposition of the amyloid precursor protein (APP) fragments with the associated appearance of degenerating cells, and blood-brain-barrier disruption. This was observed mainly in the hippocampal formation (CA2 and CA3 regions), the dentate gyrus as well as distinct regions with synaptic links to the hippocampus such as entorhinal cortex, thalamus, and basal forebrain. In addition, ET1 + Aß-treated rats also demonstrated protracted loss of AQP4 depolarization, dissolution of ß-dystroglycan, and basement membrane laminin with associated IgG and dysferlin leakage. Spatial dynamics of hippocampal injury in ET1 + Aß rats may provide a valuable model to study new targets for clinical therapeutic applications, specifically when areas remotely connected to hippocampus are damaged.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Corpo Estriado/patologia , Hipocampo/irrigação sanguínea , Hipocampo/lesões , Hipocampo/patologia , Animais , Corpo Estriado/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Ratos Wistar
9.
Sci Rep ; 8(1): 5136, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29572520

RESUMO

Ischemic stroke and diabetes are vascular risk factors for the development of impaired memory such as dementia and/or Alzheimer's disease. Clinical studies have demonstrated that minor striatal ischemic lesions in combination with ß-amyloid (Aß) load are critical in generating cognitive deficits. These cognitive deficits are likely to be associated with impaired insulin signaling. In this study, we examined the histological presence of insulin-like growth factor-I (IGF-1) and insulin receptor substrate (IRS-1) in anatomically distinct brain circuits compared with morphological brain damage in a co-morbid rat model of striatal ischemia (ET1) and Aß toxicity. The results demonstrated a rapid increase in the presence of IGF-1 and IRS-1 immunoreactive cells in Aß + ET1 rats, mainly in the ipsilateral striatum and distant regions with synaptic links to the striatal lesion. These regions included subcortical white matter, motor cortex, thalamus, dentate gyrus, septohippocampal nucleus, periventricular region and horizontal diagonal band of Broca in the basal forebrain. The alteration in IGF-1 and IRS-1 presence induced by ET1 or Aß rats alone was not severe enough to affect the entire brain circuit. Understanding the causal or etiologic interaction between insulin and IGF signaling and co-morbidity after ischemia and Aß toxicity will help design more effective therapeutics.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Isquemia Encefálica/metabolismo , Transtornos Cognitivos/metabolismo , Corpo Estriado/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Insulina/metabolismo , Transtornos da Memória/metabolismo , Transdução de Sinais , Animais , Isquemia Encefálica/patologia , Transtornos Cognitivos/patologia , Comorbidade , Corpo Estriado/patologia , Modelos Animais de Doenças , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , Transtornos da Memória/patologia , Ratos , Ratos Wistar
10.
Islets ; 10(4): 137-150, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30110202

RESUMO

Both bone marrow-derived hematopoietic stem cells (HSC) and mesenchymal stem cells (MSC) improve glycemic control in diabetic mice, but their kinetics and associated changes in pancreatic morphology have not been directly compared. Our goal was to examine the time course of improvements in glucose tolerance and associated changes in ß-cell mass and proliferation following transplantation of equivalent numbers of HSC or MSC from the same bone marrow into diabetic non-obese diabetic severe combined immune deficiency (NOD.SCID) mice. We used transgenic mice with a targeted expression of yellow fluorescent protein (YFP) driven by the Vav1 gene promoter to genetically tag HSC and progeny. HSC were separated from bone marrow by fluorescence-activated cell sorting and MSC following cell culture. Equivalent numbers of isolated HSC or MSC were transplanted directly into the pancreas of NOD.SCID mice previously made diabetic with streptozotocin. Glucose tolerance, serum insulin, ß-cell mass and ß-cell proliferation were examined up to 28 days following transplant. Transplantation with MSC improved glucose tolerance within 7 days and serum insulin levels increased, but with no increase in ß-cell mass. Mice transplanted with HSC showed improved glucose tolerance only after 3 weeks associated with increased ß-cell proliferation and mass. We conclude that single injections of either MSC or HSC transiently improved glycemic control in diabetic NOD.SCID mice, but with different time courses. However, only HSC infiltrated the islets and were associated with an expanded ß-cell mass. This suggests that MSC and HSC have differing mechanisms of action.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Hiperglicemia/terapia , Insulina/sangue , Células-Tronco Mesenquimais/fisiologia , Transplante de Células-Tronco/métodos , Animais , Glicemia/metabolismo , Medula Óssea , Modelos Animais de Doenças , Teste de Tolerância a Glucose/métodos , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fatores de Tempo , Resultado do Tratamento
11.
Islets ; 8(3): 65-82, 2016 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-27010375

RESUMO

Regeneration of insulin-producing ß-cells from resident pancreas progenitors requires an understanding of both progenitor identity and lineage plasticity. One model suggested that a rare ß-cell sub-population within islets demonstrated multi-lineage plasticity. We hypothesized that ß-cells from young mice (postnatal day 7, P7) exhibit such plasticity and used a model of islet dedifferentiation toward a ductal epithelial-cell phenotype to test this theory. RIPCre;Z/AP(+/+) mice were used to lineage trace the fate of ß-cells during dedifferentiation culture by a human placental alkaline phosphatase (HPAP) reporter. There was a significant loss of HPAP-expressing ß-cells in culture, but remaining HPAP(+) cells lost insulin expression while gaining expression of the epithelial duct cell marker cytokeratin-19 (Ck19). Flow cytometry and recovery of ß-cell subpopulations from whole pancreas vs. islets suggest that the HPAP(+)Ck19(+) cells had derived from insulin-positive, glucose-transporter-2-low (Ins(+)Glut2(LO)) cells, representing 3.5% of all insulin-expressing cells. The majority of these cells were found outside of islets within clusters of <5 ß-cells. These insulin(+)Glut2(LO) cells demonstrated a greater proliferation rate in vivo and in vitro as compared to insulin(+)Glut2(+) cells at P7, were retained into adulthood, and a subset differentiated into endocrine, ductal, and neural lineages, illustrating substantial plasticity. Results were confirmed using RIPCre;ROSA- eYFP mice. Quantitative PCR data indicated these cells possess an immature ß-cell phenotype. These Ins(+)Glut2(LO) cells may represent a resident population of cells capable of forming new, functional ß-cells, and which may be potentially exploited for regenerative therapies in the future.


Assuntos
Plasticidade Celular , Proteínas Facilitadoras de Transporte de Glucose/análise , Células Secretoras de Insulina/fisiologia , Insulina/análise , Animais , Diferenciação Celular , Linhagem da Célula/fisiologia , Citometria de Fluxo , Queratina-19/análise , Camundongos , Microscopia de Fluorescência
12.
Exp Biol Med (Maywood) ; 238(10): 1147-59, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23986224

RESUMO

Low birth weight and poor foetal growth following low protein (LP) exposure are associated with altered islet development and glucose intolerance in adulthood. Additionally, LP-fed offspring fail to regenerate their ß-cells following depletion with streptozotocin (STZ) in contrast to control-fed offspring that restore ß-cell mass. Our objective was to identify signalling pathways and cellular functions that may be critically altered in LP offspring rendering them susceptible to developing long-term glucose intolerance and decreased ß-cell plasticity. Pregnant Balb/c mice were fed a control (C; 20% protein) or an isocaloric LP (8% protein) diet throughout gestation and C diet thereafter. Female offspring were injected intraperitoneally with 35 mg/kg STZ or vehicle on days 1 to 5 for each dietary treatment. At 30 days of age, total RNA was extracted from pancreatic tissue for microarray analysis using the Affymetrix GeneChip Mouse Genome 430 2.0. Gene and protein expression were quantified from isolated islets. Finally, ß-cell proliferation was determined in vitro following REG1α treatment. The microarray data and GO enrichment analysis indicated that foetal protein restriction alters the early expression of genes necessary for many cell functions, such as oxidative phosphorylation and free radical scavenging. Expression of Reg1 was upregulated following STZ, whereas protein content was decreased in LP + STZ islets. Furthermore, REG1α failed to stimulate ß-cell proliferation in vitro in LP + STZ islets. Therefore, early nutritional insults may programme the Reg1 pathway resulting in a limited ability to increase ß-cell mass during metabolic stress. In conclusion, this study implicates the Reg1 pathway in ß-cell regeneration and describes altered programming of gene expression in LP offspring, which underlies later development of cell dysfunction and glucose intolerance in adulthood.


Assuntos
Proliferação de Células , Intolerância à Glucose/fisiopatologia , Células Secretoras de Insulina/fisiologia , Litostatina/metabolismo , Deficiência de Proteína/complicações , Regeneração , Irmãos , Animais , Feminino , Perfilação da Expressão Gênica , Litostatina/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Gravidez , Proteínas , Proteoma/análise , Estreptozocina/administração & dosagem , Estreptozocina/toxicidade
13.
Exp Biol Med (Maywood) ; 235(2): 189-98, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20404034

RESUMO

Neonatal rats undergo considerable beta-cell regeneration after depletion with streptozotocin (STZ). Since the intraislet vasculature is necessary for both beta-cell growth and function, we examined changes in vascular morphology following STZ. Neonatal Wistar rats (4 days) were injected with 70 mg/kg STZ, or buffer, and were examined between days 4 and 40 postinjection. Animals receiving STZ were relatively hyperglycemic for eight days, but became normoglycemic subsequent to a partial recovery of beta-cell mass. However, glucose tolerance remained impaired. The intraislet area occupied by capillaries was significantly reduced by approximately 20% following STZ, mainly within the beta-cell-rich islet core, but had recovered by day 40. Vascular endothelial growth factor (VEGF) was localized to beta-cells, and pancreatic VEGF mRNA levels in control animals showed a progressive increase between days 4 and 20. This rise was delayed following STZ, but by day 20 VEGF mRNA abundance exceeded that in control pancreas. Hepatocyte growth factor (HGF) was localized to intraislet endothelial cells. Levels of HGF mRNA increased until day 16 in control rats, but subsequently declined to low levels. Following STZ, HGF mRNA had declined prematurely after day 12. Type IV collagen was localized to the extracellular matrix around the intraislet vasculature. The islet area immunopositive for collagen was significantly reduced at all times following STZ. Results indicate that there is a relative loss of intraislet vasculature following STZ, which may limit subsequent beta-cell regeneration through both local growth factor and extracellular matrix interactions.


Assuntos
Diabetes Mellitus Experimental/patologia , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/patologia , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Colágeno Tipo IV/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Regeneração , Estreptozocina/toxicidade , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
J Endocrinol ; 201(2): 231-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19228796

RESUMO

Dietary insult in early life can affect the development and future function of the endocrine pancreas. We maintained pregnant non-obese diabetic (NOD) mice on a low protein (LP, 8% protein versus control, 20%) diet from conception until the weaning of pups at day 21. Serum insulin and pancreatic insulin content were reduced in LP-fed NOD offspring at 8 weeks, as were serum interferon gamma and pancreatic tumor necrosis factor alpha, while the number of pancreatic islets demonstrating peri-insulitis, and the degree of invasiveness were reduced. To determine if LP caused early morphometric changes in the pancreas, we measured mean islet area at days 3 and 21. Mean islet size did not differ with diet, but by 8 weeks of age LP-fed NOD females exhibited a significantly reduced islet number and mean islet area, and a lower fractional area of pancreas occupied by both alpha- and beta-cells than control-fed mice. The onset of diabetes was delayed in NOD mice of both genders fed LP diet. The mechanism is likely to involve both altered beta-cell morphology and function and changes in cytotoxic cytokines.


Assuntos
Diabetes Mellitus Tipo 1/patologia , Dieta com Restrição de Proteínas , Camundongos Endogâmicos NOD , Fatores Etários , Idade de Início , Ração Animal , Animais , Animais Recém-Nascidos , Diabetes Mellitus Tipo 1/epidemiologia , Feminino , Insulina/sangue , Tamanho da Ninhada de Vivíparos , Masculino , Camundongos , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA