Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Vis Exp ; (191)2023 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-37602849

RESUMO

ARTICLES DISCUSSED: McCarthy, E., Rowe, C. J., Crowley-Perry, M., Connaughton, V. P. Alternate immersion in glucose to produce prolonged hyperglycemia in zebrafish. Journal of Visualized Experiments. (171), e61935 (2021). Rowe, C. J., Crowley-Perry, M., McCarthy, E., Davidson, T. L., Connaughton, V. P. The three-chamber choice behavioral task using zebrafish as a model system. Journal of Visualized Experiments. (170), e61934 (2021). Memon, B., Abdelalim, E. M. Differentiation of human pluripotent stem cells into pancreatic beta-cell precursors in a 2D culture system. Journal of Visualized Experiments. (178), e63298 (2021). Aghadi, M., Karam, M., Abdelalim, E. M. Robust differentiation of human iPSCs into a pure population of adipocytes to study adipocyte-associated disorders. Journal of Visualized Experiments. (180), e63311 (2022). Galmozzi, A., Kok, B. P., Saez, E. Isolation and differentiation of primary white and brown preadipocytes from newborn mice. Journal of Visualized Experiments. (167), e62005 (2021). Pérez Gutiérrez, R. M., Arrioja, M. W. Rapid model to evaluate the anti-obesity potential of a combination of Syzygium aromaticum (clove) and Cuminum cyminum (cumin) on C57BL6/j mice fed high-fat diet. Journal of Visualized Experiments. (173), e62087 (2021).


Assuntos
Diabetes Mellitus , Hiperglicemia , Humanos , Animais , Camundongos , Peixe-Zebra , Adipócitos , Diferenciação Celular , Camundongos Endogâmicos C57BL , Obesidade
2.
Methods Cell Biol ; 175: 177-219, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36967141

RESUMO

In this chapter we provide some tools to study the ciliary proteins that make it possible for Paramecium cells to swim by beating their cilia. These proteins include many ion channels, accessory proteins, peripheral proteins, structural proteins, rootlets of cilia, and enzymes. Some of these proteins are also found in the soma membrane, but their distinct and critical functions are in the cilia. Paramecium has 4000 or more cilia per cell, giving it an advantage for biochemical studies over cells that have one primarily cilium per cell. Nonetheless, a challenge for studies of many ciliary proteins in Paramecium is their low abundance. We discuss here several strategies to overcome this challenge and other challenges such as working with very large channel proteins. We also include for completeness other techniques that are critical to the study of swimming behavior, such as genetic crosses, recording of swimming patterns, electrical recordings, expression of very large channel proteins, RNA Interference, among others.


Assuntos
Paramecium tetraurellia , Paramecium , Paramecium tetraurellia/genética , Paramecium tetraurellia/metabolismo , Cílios/metabolismo , Paramecium/genética , Paramecium/metabolismo , Proteínas de Membrana/metabolismo
3.
Metabolites ; 11(8)2021 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-34436454

RESUMO

Transcription factor 19 (TCF19) is a gene associated with type 1 diabetes (T1DM) and type 2 diabetes (T2DM) in genome-wide association studies. Prior studies have demonstrated that Tcf19 knockdown impairs ß-cell proliferation and increases apoptosis. However, little is known about its role in diabetes pathogenesis or the effects of TCF19 gain-of-function. The aim of this study was to examine the impact of TCF19 overexpression in INS-1 ß-cells and human islets on proliferation and gene expression. With TCF19 overexpression, there was an increase in nucleotide incorporation without any change in cell cycle gene expression, alluding to an alternate process of nucleotide incorporation. Analysis of RNA-seq of TCF19 overexpressing cells revealed increased expression of several DNA damage response (DDR) genes, as well as a tightly linked set of genes involved in viral responses, immune system processes, and inflammation. This connectivity between DNA damage and inflammatory gene expression has not been well studied in the ß-cell and suggests a novel role for TCF19 in regulating these pathways. Future studies determining how TCF19 may modulate these pathways can provide potential targets for improving ß-cell survival.

4.
Yale J Biol Med ; 92(3): 471-480, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31543709

RESUMO

A primary challenge in type 2 diabetes (T2D) is the preservation of a functional population of ß-cells, which play a central role in regulating blood glucose levels. Two congenital disorders, Bardet-Biedl syndrome (BBS) and Alström syndrome (ALMS), can serve as useful models to understand how ß-cells are normally produced and regenerated. Both are characterized by obesity, loss of ß-cells, and defects in primary cilia - the sensory center of cells. Primary cilia are cellular protrusions present in almost every vertebrate cell. This antenna-like organelle plays a crucial role in regulating several signaling pathways that direct proper development, proliferation, and homeostasis. Mutations in genes expressing ciliary proteins or proteins present at or near the base of the cilium lead to disorders, collectively called ciliopathies. BBS and Alström syndrome are such disorders. Though both BBS and Alström patients are obese, their childhood diabetes rates are vastly different, suggesting distinct pathogenesis underlying these two ciliopathies. Clinical studies suggest that BBS patients are protected against early onset diabetes by sustained or enhanced ß-cell function. In contrast, Alström patients are more prone to develop diabetes. They have hyperinsulinemia, yet their ß-cells fail to sense glucose and to regulate insulin secretion accordingly. These data suggest a potential role for primary cilia in maintaining a functional ß-cell population and that defects in cilia or in ciliary proteins impair development and function of ß-cells. Identifying the respective roles of primary cilia and ciliary proteins, such as BBS and ALMS1 may shed light on ß-cell biology and uncover potentially novel targets for diabetes therapy.


Assuntos
Cílios/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Ciliopatias/metabolismo , Humanos , Insulina/biossíntese , Modelos Biológicos
5.
Hum Mol Genet ; 28(13): 2212-2223, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31220269

RESUMO

Alström syndrome (OMIM #203800) is an autosomal recessive obesity ciliopathy caused by loss-of-function mutations in the ALMS1 gene. In addition to multi-organ dysfunction, such as cardiomyopathy, retinal degeneration and renal dysfunction, the disorder is characterized by high rates of obesity, insulin resistance and early-onset type 2 diabetes mellitus (T2DM). To investigate the underlying mechanisms of T2DM phenotypes, we generated a loss-of-function deletion of alms1 in the zebrafish. We demonstrate conservation of hallmark clinical characteristics alongside metabolic syndrome phenotypes, including a propensity for obesity and fatty livers, hyperinsulinemia and glucose response defects. Gene expression changes in ß-cells isolated from alms1-/- mutants revealed changes consistent with insulin hypersecretion and glucose sensing failure, which were corroborated in cultured murine ß-cells lacking Alms1. We also found evidence of defects in peripheral glucose uptake and concomitant hyperinsulinemia in the alms1-/- animals. We propose a model in which hyperinsulinemia is the primary and causative defect underlying generation of T2DM associated with alms1 deficiency. These observations support the alms1 loss-of-function zebrafish mutant as a monogenic model for mechanistic interrogation of T2DM phenotypes.


Assuntos
Síndrome de Alstrom/genética , Diabetes Mellitus Tipo 2/genética , Resistência à Insulina/genética , Insuficiência Renal/genética , Degeneração Retiniana/genética , Peixe-Zebra/genética , Síndrome de Alstrom/fisiopatologia , Animais , Animais Geneticamente Modificados , Linhagem Celular , Modelos Animais de Doenças , Intolerância à Glucose , Hiperinsulinismo/genética , Células Secretoras de Insulina/metabolismo , Camundongos , Modelos Biológicos , Obesidade/genética , Fenótipo , Peixe-Zebra/embriologia
6.
Sci Rep ; 7(1): 9765, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28852127

RESUMO

Bardet-Biedl syndrome is a model ciliopathy. Although the characterization of BBS proteins has evidenced their involvement in cilia, extraciliary functions for some of these proteins are also being recognized. Importantly, understanding both cilia and cilia-independent functions of the BBS proteins is key to fully dissect the cellular basis of the syndrome. Here we characterize a functional interaction between BBS4 and the secreted protein FSTL1, a protein linked to adipogenesis and inflammation among other functions. We show that BBS4 and cilia regulate FSTL1 mRNA levels, but BBS4 also modulates FSTL1 secretion. Moreover, we show that FSTL1 is a novel regulator of ciliogenesis thus underscoring a regulatory loop between FSTL1 and cilia. Finally, our data indicate that BBS4, cilia and FSTL1 are coordinated during the differentiation of 3T3-L1 cells and that FSTL1 plays a role in this process, at least in part, by modulating ciliogenesis. Therefore, our findings are relevant to fully understand the development of BBS-associated phenotypes such as obesity.


Assuntos
Diferenciação Celular/genética , Cílios/genética , Cílios/metabolismo , Proteínas Relacionadas à Folistatina/biossíntese , Proteínas Relacionadas à Folistatina/genética , Regulação da Expressão Gênica , Proteínas/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/genética , Animais , Técnicas de Silenciamento de Genes , Espaço Intracelular/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos , Proteínas/genética
7.
J Exp Biol ; 219(Pt 19): 3028-3038, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27707864

RESUMO

Paramecium cells swim by beating their cilia, and make turns by transiently reversing their power stroke. Reversal is caused by Ca2+ entering the cilium through voltage-gated Ca2+ (CaV) channels that are found exclusively in the cilia. As ciliary Ca2+ levels return to normal, the cell pivots and swims forward in a new direction. Thus, the activation of the CaV channels causes cells to make a turn in their swimming paths. For 45 years, the physiological characteristics of the Paramecium ciliary CaV channels have been known, but the proteins were not identified until recently, when the P. tetraurelia ciliary membrane proteome was determined. Three CaVα1 subunits that were identified among the proteins were cloned and confirmed to be expressed in the cilia. We demonstrate using RNA interference that these channels function as the ciliary CaV channels that are responsible for the reversal of ciliary beating. Furthermore, we show that Pawn (pw) mutants of Paramecium that cannot swim backward for lack of CaV channel activity do not express any of the three CaV1 channels in their ciliary membrane, until they are rescued from the mutant phenotype by expression of the wild-type PW gene. These results reinforce the correlation of the three CaV channels with backward swimming through ciliary reversal. The PwB protein, found in endoplasmic reticulum fractions, co-immunoprecipitates with the CaV1c channel and perhaps functions in trafficking. The PwA protein does not appear to have an interaction with the channel proteins but affects their appearance in the cilia.


Assuntos
Canais de Cálcio/metabolismo , Cílios/metabolismo , Paramecium/metabolismo , Sequência de Bases , Western Blotting , Canais de Cálcio/química , Sequência Consenso , Imunoprecipitação , Mutação/genética , Fenótipo , Ligação Proteica , Domínios Proteicos , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Coloração pela Prata , Soluções , Frações Subcelulares/metabolismo , Natação
8.
BMC Genomics ; 17: 318, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27142762

RESUMO

BACKGROUND: Bardet-Biedl Syndrome (BBS) and Alström Syndrome are two pleiotropic ciliopathies with significant phenotypic overlap between them across many tissues. Although BBS and Alström genes are necessary for the proper function of primary cilia, their role in defects across multiple organ systems is unclear. METHODS: To provide insight into the pathways underlying BBS and Alström phenotypes, we carried out whole organism transcriptome analysis by RNA sequencing in established zebrafish models of the syndromes. RESULTS: We analyzed all genes that were significantly differentially expressed and found enrichment of phenotypically significant pathways in both models. These included multiple pathways shared between the two disease models as well as those unique to each model. Notably, we identified significant downregulation of genes in pathways relevant to visual system deficits and obesity in both disorders, consistent with those shared phenotypes. In contrast, neuronal pathways were significantly downregulated only in the BBS model but not in the Alström model. Our observations also suggested an important role for G-protein couple receptor and calcium signaling defects in both models. DISCUSSION: Pathway network analyses of both models indicate that visual system defects may be driven by genetic mechanisms independent of other phenotypes whereas the majority of other phenotypes are a result of genetic players that contribute to multiple pathways simultaneously. Additionally, examination of genes differentially expressed in opposing directions between the two models suggest a deficit in pancreatic function in the Alström model, that is not present in the BBS model. CONCLUSIONS: These findings provide important novel insight into shared and divergent phenotypes between two similar but distinct genetic syndromes.


Assuntos
Síndrome de Alstrom/genética , Síndrome de Bardet-Biedl/genética , Perfilação da Expressão Gênica , Fenótipo , Transcriptoma , Peixe-Zebra/genética , Síndrome de Alstrom/diagnóstico , Animais , Síndrome de Bardet-Biedl/diagnóstico , Biologia Computacional/métodos , Modelos Animais de Doenças , Regulação da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Transdução de Sinal Luminoso , Vias Neurais , Vias Visuais
9.
Hum Mol Genet ; 25(1): 57-68, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26494903

RESUMO

Rare genetic syndromes characterized by early-onset type 2 diabetes have revealed the importance of pancreatic ß-cells in genetic susceptibility to diabetes. However, the role of genetic regulation of ß-cells in disorders that are also characterized by highly penetrant obesity, a major additional risk factor, is unclear. In this study, we investigated the contribution of genes associated with two obesity ciliopathies, Bardet-Biedl Syndrome and Alstrom Syndrome, to the production and maintenance of pancreatic ß-cells. Using zebrafish models of these syndromes, we identified opposing effects on production of ß-cells. Loss of the Alstrom gene, alms1, resulted in a significant decrease in ß-cell production whereas loss of BBS genes, bbs1 or bbs4, resulted in a significant increase. Examination of the regulatory program underlying ß-cell production suggested that these effects were specific to ß-cells. In addition to the initial production of ß-cells, we observed significant differences in their continued maintenance. Under prolonged exposure to high glucose conditions, alms1-deficient ß-cells were unable to continually expand as a result of decreased proliferation and increased cell death. Although bbs1-deficient ß-cells were similarly susceptible to apoptosis, the overall maintenance of ß-cell number in those animals was sustained likely due to increased proliferation. Taken together, these findings implicate discrepant production and maintenance of ß-cells in the differential susceptibility to diabetes found between these two genetic syndromes.


Assuntos
Síndrome de Alstrom/genética , Síndrome de Bardet-Biedl/genética , Células Secretoras de Insulina/patologia , Animais , Morte Celular , Proliferação de Células , Modelos Animais de Doenças , Glucose , Hiperglicemia/patologia , Proteínas Associadas aos Microtúbulos/genética , Morfolinos/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
10.
Birth Defects Res C Embryo Today ; 102(2): 139-58, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24864023

RESUMO

Primary cilia and their anchoring basal bodies are important regulators of a growing list of signaling pathways. Consequently, dysfunction in proteins associated with these structures results in perturbation of the development and function of a spectrum of tissue and cell types. Here, we review the role of cilia in mediating the development and function of the pancreas. We focus on ciliary regulation of major pathways involved in pancreatic development, including Shh, Wnt, TGF-ß, Notch, and fibroblast growth factor. We also discuss pancreatic phenotypes associated with ciliary dysfunction, including pancreatic cysts and defects in glucose homeostasis, and explore the potential role of cilia in such defects.


Assuntos
Cílios/patologia , Organogênese/genética , Pâncreas/patologia , Animais , Modelos Animais de Doenças , Transtornos do Metabolismo de Glucose/genética , Transtornos do Metabolismo de Glucose/patologia , Humanos , Pâncreas/citologia , Pâncreas/metabolismo , Cisto Pancreático/genética , Cisto Pancreático/patologia , Fenótipo , Transdução de Sinais
11.
J Cell Sci ; 127(Pt 11): 2407-19, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24681783

RESUMO

Proteins associated with primary cilia and basal bodies mediate numerous signaling pathways, but little is known about their role in Notch signaling. Here, we report that loss of the Bardet-Biedl syndrome proteins BBS1 or BBS4 produces increased Notch-directed transcription in a zebrafish reporter line and in human cell lines. Pathway overactivation is accompanied by reduced localization of Notch receptor at both the plasma membrane and the cilium. In Drosophila mutants, overactivation of Notch can result from receptor accumulation in endosomes, and recent studies implicate ciliary proteins in endosomal trafficking, suggesting a possible mechanism by which overactivation occurs in BBS mutants. Consistent with this, we observe genetic interaction of BBS1 and BBS4 with the endosomal sorting complexes required for transport (ESCRT) gene TSG101 and accumulation of receptor in late endosomes, reduced endosomal recycling and reduced receptor degradation in lysosomes. We observe similar defects with disruption of BBS3. Loss of another basal body protein, ALMS1, also enhances Notch activation and the accumulation of receptor in late endosomes, but does not disrupt recycling. These findings suggest a role for these proteins in the regulation of Notch through endosomal trafficking of the receptor.


Assuntos
Corpos Basais/fisiologia , Membrana Celular/metabolismo , Cílios/fisiologia , Endossomos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas/metabolismo , Receptores Notch/metabolismo , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/genética , Mutação/genética , Transporte Proteico/genética , Proteínas/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...