Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Diabetologia ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38743124

RESUMO

AIMS/HYPOTHESIS: Regulatory factor X 6 (RFX6) is crucial for pancreatic endocrine development and differentiation. The RFX6 variant p.His293LeufsTer7 is significantly enriched in the Finnish population, with almost 1:250 individuals as a carrier. Importantly, the FinnGen study indicates a high predisposition for heterozygous carriers to develop type 2 and gestational diabetes. However, the precise mechanism of this predisposition remains unknown. METHODS: To understand the role of this variant in beta cell development and function, we used CRISPR technology to generate allelic series of pluripotent stem cells. We created two isogenic stem cell models: a human embryonic stem cell model; and a patient-derived stem cell model. Both were differentiated into pancreatic islet lineages (stem-cell-derived islets, SC-islets), followed by implantation in immunocompromised NOD-SCID-Gamma mice. RESULTS: Stem cell models of the homozygous variant RFX6-/- predictably failed to generate insulin-secreting pancreatic beta cells, mirroring the phenotype observed in Mitchell-Riley syndrome. Notably, at the pancreatic endocrine stage, there was an upregulation of precursor markers NEUROG3 and SOX9, accompanied by increased apoptosis. Intriguingly, heterozygous RFX6+/- SC-islets exhibited RFX6 haploinsufficiency (54.2% reduction in protein expression), associated with reduced beta cell maturation markers, altered calcium signalling and impaired insulin secretion (62% and 54% reduction in basal and high glucose conditions, respectively). However, RFX6 haploinsufficiency did not have an impact on beta cell number or insulin content. The reduced insulin secretion persisted after in vivo implantation in mice, aligning with the increased risk of variant carriers to develop diabetes. CONCLUSIONS/INTERPRETATION: Our allelic series isogenic SC-islet models represent a powerful tool to elucidate specific aetiologies of diabetes in humans, enabling the sensitive detection of aberrations in both beta cell development and function. We highlight the critical role of RFX6 in augmenting and maintaining the pancreatic progenitor pool, with an endocrine roadblock and increased cell death upon its loss. We demonstrate that RFX6 haploinsufficiency does not affect beta cell number or insulin content but does impair function, predisposing heterozygous carriers of loss-of-function variants to diabetes. DATA AVAILABILITY: Ultra-deep bulk RNA-seq data for pancreatic differentiation stages 3, 5 and 7 of H1 RFX6 genotypes are deposited in the Gene Expression Omnibus database with accession code GSE234289. Original western blot images are deposited at Mendeley ( https://data.mendeley.com/datasets/g75drr3mgw/2 ).

2.
Diabetes ; 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38603470

RESUMO

Pluripotent stem cell-derived islets (SC-islets) now emerge as a new source for beta-cell replacement therapy. While the function of human islet transplants is hampered by excessive cell death post-transplantation, contributing factors include inflammatory reactions, insufficient revascularization and islet amyloid formation, there is a gap in knowledge on the engraftment process of the SC-islets. In this experimental study, we investigated the engraftment capability of SC-islets at three months post-transplantation, and observed that the cell apoptosis rates were lower, but the vascular density was similar in SC-islets to that of human islets. While the human islet transplant vascular structures were a mixture of remnant donor endothelium and ingrowing blood vessels, the SC-islets contained ingrowing blood vessels only. The oxygenation of the SC-islet grafts was twice as high as in the corresponding grafts of human islets, suggesting better vascular functionality. Similar to the blood vessel ingrowth, also the reinnervation of the SC-islets was four- to five-fold higher than the human islets. Both SC-islets and the human islets contained amyloid at one and three months post-transplantation. We conclude that the vascular and neural engraftment of SC-islets is superior to human islets, but that grafts of both origins develop amyloid with potential long-term consequences.

3.
Nat Genet ; 55(12): 2075-2081, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37973953

RESUMO

Identifying genes linked to extreme phenotypes in humans has the potential to highlight biological processes not shared with all other mammals. Here, we report the identification of homozygous loss-of-function variants in the primate-specific gene ZNF808 as a cause of pancreatic agenesis. ZNF808 is a member of the KRAB zinc finger protein family, a large and rapidly evolving group of epigenetic silencers which target transposable elements. We show that loss of ZNF808 in vitro results in aberrant activation of regulatory potential contained in the primate-specific transposable elements it represses during early pancreas development. This leads to inappropriate specification of cell fate with induction of genes associated with liver identity. Our results highlight the essential role of ZNF808 in pancreatic development in humans and the contribution of primate-specific regions of the human genome to congenital developmental disease.


Assuntos
Anormalidades Congênitas , Elementos de DNA Transponíveis , Proteínas de Ligação a DNA , Pâncreas , Animais , Humanos , Diferenciação Celular , Genoma Humano , Primatas/anormalidades , Primatas/genética , Proteínas de Ligação a DNA/genética , Anormalidades Congênitas/genética , Pâncreas/anormalidades
4.
Cell Rep ; 42(8): 112970, 2023 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-37556323

RESUMO

Pancreatic islets regulate blood glucose homeostasis through the controlled release of insulin; however, current metabolic models of glucose-sensitive insulin secretion are incomplete. A comprehensive understanding of islet metabolism is integral to studies of endocrine cell development as well as diabetic islet dysfunction. Human pluripotent stem cell-derived islets (SC-islets) are a developmentally relevant model of human islet function that have great potential in providing a cure for type 1 diabetes. Using multiple 13C-labeled metabolic fuels, we demonstrate that SC-islets show numerous divergent patterns of metabolite trafficking in proposed insulin release pathways compared with primary human islets but are still reliant on mitochondrial aerobic metabolism to derive function. Furthermore, reductive tricarboxylic acid cycle activity and glycolytic metabolite cycling occur in SC-islets, suggesting that non-canonical coupling factors are also present. In aggregate, we show that many facets of SC-islet metabolism overlap with those of primary islets, albeit with a retained immature signature.

5.
Nat Commun ; 13(1): 6363, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36289205

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease that results in the destruction of insulin producing pancreatic ß-cells. One of the genes associated with T1D is TYK2, which encodes a Janus kinase with critical roles in type-Ι interferon (IFN-Ι) mediated intracellular signalling. To study the role of TYK2 in ß-cell development and response to IFNα, we generated TYK2 knockout human iPSCs and directed them into the pancreatic endocrine lineage. Here we show that loss of TYK2 compromises the emergence of endocrine precursors by regulating KRAS expression, while mature stem cell-islets (SC-islets) function is not affected. In the SC-islets, the loss or inhibition of TYK2 prevents IFNα-induced antigen processing and presentation, including MHC Class Ι and Class ΙΙ expression, enhancing their survival against CD8+ T-cell cytotoxicity. These results identify an unsuspected role for TYK2 in ß-cell development and support TYK2 inhibition in adult ß-cells as a potent therapeutic target to halt T1D progression.


Assuntos
Diabetes Mellitus Tipo 1 , Insulinas , Humanos , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Insulinas/metabolismo , Interferon-alfa/farmacologia , Interferon-alfa/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , TYK2 Quinase/genética , TYK2 Quinase/metabolismo , Células Secretoras de Insulina
6.
STAR Protoc ; 3(4): 101711, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36136756

RESUMO

We present here a robust and reliable protocol by which to differentiate pancreatic islet-like aggregates (SC-islets) from human pluripotent stem cells. The 7-stage protocol mimics developmental patterning factors that induce endocrine lineage formation and spans monolayer, microwell, and aggregate suspension culture. The SC-islets demonstrate dynamic glucose-sensitive insulin secretion and an endocrine cell composition similar to those of primary human islets. SC-islets generated using this optimized protocol are suitable for in vitro modeling of islet cell pathophysiology and therapeutic applications. For complete details on the use and execution of this protocol, please refer to Balboa et al. (2022).


Assuntos
Ilhotas Pancreáticas , Células-Tronco Pluripotentes , Humanos , Diferenciação Celular/fisiologia , Glucose/metabolismo , Secreção de Insulina
7.
Nat Biotechnol ; 40(7): 1042-1055, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35241836

RESUMO

Transplantation of pancreatic islet cells derived from human pluripotent stem cells is a promising treatment for diabetes. Despite progress in the generation of stem-cell-derived islets (SC-islets), no detailed characterization of their functional properties has been conducted. Here, we generated functionally mature SC-islets using an optimized protocol and benchmarked them comprehensively against primary adult islets. Biphasic glucose-stimulated insulin secretion developed during in vitro maturation, associated with cytoarchitectural reorganization and the increasing presence of alpha cells. Electrophysiology, signaling and exocytosis of SC-islets were similar to those of adult islets. Glucose-responsive insulin secretion was achieved despite differences in glycolytic and mitochondrial glucose metabolism. Single-cell transcriptomics of SC-islets in vitro and throughout 6 months of engraftment in mice revealed a continuous maturation trajectory culminating in a transcriptional landscape closely resembling that of primary islets. Our thorough evaluation of SC-islet maturation highlights their advanced degree of functionality and supports their use in further efforts to understand and combat diabetes.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Células-Tronco Pluripotentes , Animais , Glucose/metabolismo , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Camundongos , Células-Tronco Pluripotentes/metabolismo
8.
Diabetes ; 70(4): 1006-1018, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33500254

RESUMO

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-resident protein that plays a crucial role in attenuating ER stress responses. Although MANF is indispensable for the survival and function of mouse ß-cells, its precise role in human ß-cell development and function is unknown. In this study, we show that lack of MANF in humans results in diabetes due to increased ER stress, leading to impaired ß-cell function. We identified two patients from different families with childhood diabetes and a neurodevelopmental disorder associated with homozygous loss-of-function mutations in the MANF gene. To study the role of MANF in human ß-cell development and function, we knocked out the MANF gene in human embryonic stem cells and differentiated them into pancreatic endocrine cells. Loss of MANF induced mild ER stress and impaired insulin-processing capacity of ß-cells in vitro. Upon implantation to immunocompromised mice, the MANF knockout grafts presented elevated ER stress and functional failure, particularly in recipients with diabetes. By describing a new form of monogenic neurodevelopmental diabetes syndrome caused by disturbed ER function, we highlight the importance of adequate ER stress regulation for proper human ß-cell function and demonstrate the crucial role of MANF in this process.


Assuntos
Estresse do Retículo Endoplasmático/genética , Fatores de Crescimento Neural/metabolismo , Western Blotting , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Edição de Genes/métodos , Teste de Tolerância a Glucose , Humanos , Imuno-Histoquímica , Masculino , Mutação/genética , Fatores de Crescimento Neural/genética , Reação em Cadeia da Polimerase em Tempo Real , Estreptozocina/farmacologia
9.
Diabetologia ; 64(3): 630-640, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33404684

RESUMO

AIMS/HYPOTHESIS: Congenital hyperinsulinism caused by mutations in the KATP-channel-encoding genes (KATPHI) is a potentially life-threatening disorder of the pancreatic beta cells. No optimal medical treatment is available for patients with diazoxide-unresponsive diffuse KATPHI. Therefore, we aimed to create a model of KATPHI using patient induced pluripotent stem cell (iPSC)-derived islets. METHODS: We derived iPSCs from a patient carrying a homozygous ABCC8V187D mutation, which inactivates the sulfonylurea receptor 1 (SUR1) subunit of the KATP-channel. CRISPR-Cas9 mutation-corrected iPSCs were used as controls. Both were differentiated to stem cell-derived islet-like clusters (SC-islets) and implanted into NOD-SCID gamma mice. RESULTS: SUR1-mutant and -corrected iPSC lines both differentiated towards the endocrine lineage, but SUR1-mutant stem cells generated 32% more beta-like cells (SC-beta cells) (64.6% vs 49.0%, p = 0.02) and 26% fewer alpha-like cells (16.1% vs 21.8% p = 0.01). SUR1-mutant SC-beta cells were 61% more proliferative (1.23% vs 0.76%, p = 0.006), and this phenotype could be induced in SUR1-corrected cells with pharmacological KATP-channel inactivation. The SUR1-mutant SC-islets secreted 3.2-fold more insulin in low glucose conditions (0.0174% vs 0.0054%/min, p = 0.0021) and did not respond to KATP-channel-acting drugs in vitro. Mice carrying grafts of SUR1-mutant SC-islets presented with 38% lower fasting blood glucose (4.8 vs 7.7 mmol/l, p = 0.009) and their grafts failed to efficiently shut down insulin secretion during induced hypoglycaemia. Explanted SUR1-mutant grafts displayed an increase in SC-beta cell proportion and SC-beta cell nucleomegaly, which was independent of proliferation. CONCLUSIONS/INTERPRETATION: We have created a model recapitulating the known pathophysiology of KATPHI both in vitro and in vivo. We have also identified a novel role for KATP-channel activity during human islet development. This model will enable further studies for the improved understanding and clinical management of KATPHI without the need for primary patient tissue.


Assuntos
Hiperinsulinismo Congênito/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores de Sulfonilureias/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Hiperinsulinismo Congênito/genética , Hiperinsulinismo Congênito/patologia , Hiperinsulinismo Congênito/fisiopatologia , Feminino , Predisposição Genética para Doença , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/transplante , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Mutação , Fenótipo , Receptores de Sulfonilureias/genética
10.
J Clin Invest ; 130(12): 6338-6353, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33164986

RESUMO

Neonatal diabetes is caused by single gene mutations reducing pancreatic ß cell number or impairing ß cell function. Understanding the genetic basis of rare diabetes subtypes highlights fundamental biological processes in ß cells. We identified 6 patients from 5 families with homozygous mutations in the YIPF5 gene, which is involved in trafficking between the endoplasmic reticulum (ER) and the Golgi. All patients had neonatal/early-onset diabetes, severe microcephaly, and epilepsy. YIPF5 is expressed during human brain development, in adult brain and pancreatic islets. We used 3 human ß cell models (YIPF5 silencing in EndoC-ßH1 cells, YIPF5 knockout and mutation knockin in embryonic stem cells, and patient-derived induced pluripotent stem cells) to investigate the mechanism through which YIPF5 loss of function affects ß cells. Loss of YIPF5 function in stem cell-derived islet cells resulted in proinsulin retention in the ER, marked ER stress, and ß cell failure. Partial YIPF5 silencing in EndoC-ßH1 cells and a patient mutation in stem cells increased the ß cell sensitivity to ER stress-induced apoptosis. We report recessive YIPF5 mutations as the genetic cause of a congenital syndrome of microcephaly, epilepsy, and neonatal/early-onset diabetes, highlighting a critical role of YIPF5 in ß cells and neurons. We believe this is the first report of mutations disrupting the ER-to-Golgi trafficking, resulting in diabetes.


Assuntos
Diabetes Mellitus , Estresse do Retículo Endoplasmático/genética , Doenças Genéticas Inatas , Doenças do Recém-Nascido , Microcefalia , Mutação , Proteínas de Transporte Vesicular , Linhagem Celular , Diabetes Mellitus/embriologia , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Feminino , Doenças Genéticas Inatas/embriologia , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/patologia , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Recém-Nascido , Doenças do Recém-Nascido/embriologia , Doenças do Recém-Nascido/genética , Doenças do Recém-Nascido/patologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Microcefalia/embriologia , Microcefalia/genética , Microcefalia/patologia , Neurônios/metabolismo , Neurônios/patologia , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
11.
Ups J Med Sci ; 124(4): 228-237, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31623497

RESUMO

Background: Murine boundary cap-derived neural crest stem cells (NCSCs) are capable of enhancing islet function by stimulating beta cell proliferation as well as increasing the neural and vascular density in the islets both in vitro and in vivo. This study aimed to isolate NCSC-like cells from human bone marrow.Methods: CD271 magnetic cell separation and culture techniques were used to purify a NCSC-enriched population of human bone marrow. Analyses of the CD271+ and CD271- fractions in terms of protein expression were performed, and the capacity of the CD271+ bone marrow cells to form 3-dimensional spheres when grown under non-adherent conditions was also investigated. Moreover, the NCSC characteristics of the CD271+ cells were evaluated by their ability to migrate toward human islets as well as human islet-like cell clusters (ICC) derived from pluripotent stem cells.Results: The CD271+ bone marrow population fulfilled the criterion of being multipotent stem cells, having the potential to differentiate into glial cells, neurons as well as myofibroblasts in vitro. They had the capacity to form 3-dimensional spheres as well as an ability to migrate toward human islets, further supporting their NCSC identity. Additionally, we demonstrated similar migration features toward stem cell-derived ICC.Conclusion: The results support the NCSC identity of the CD271-enriched human bone marrow population. It remains to investigate whether the human bone marrow-derived NCSCs have the ability to improve transplantation efficacy of not only human islets but stem cell-derived ICC as well.


Assuntos
Técnicas de Cultura de Células , Separação Celular/métodos , Ilhotas Pancreáticas/citologia , Crista Neural/citologia , Células-Tronco Pluripotentes/citologia , Adapaleno/metabolismo , Adulto , Idoso , Células da Medula Óssea/citologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Humanos , Transplante das Ilhotas Pancreáticas , Masculino , Pessoa de Meia-Idade , Adulto Jovem
12.
Stem Cells ; 37(1): 33-41, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30270471

RESUMO

Pancreatic ß-cells are the only source of insulin. Disturbances in ß-cell development or function may thus result in insulin deficiency or excess, presenting as hyper- or hypoglycemia. It is increasingly evident that common forms of diabetes (types 1 and 2) are pathogenically heterogeneous. Development of efficient therapies is dependent on reliable disease models. Although animal models are remarkably useful research tools, they present limitations because of species differences. As an alternative, human pluripotent stem cell technologies offer multiple possibilities for the study of human diseases in vitro. In the last decade, advances in the derivation of induced pluripotent stem cells from diabetic patients, combined with ß-cell differentiation protocols, have resulted in the generation of useful disease models for diabetes. First disease models have been focusing on monogenic diabetes. The development of genome editing technologies, more advanced differentiation protocols and humanized mouse models based on transplanted cells have opened new horizons for the modeling of more complex forms of ß-cell dysfunction. We present here the incremental progress made in the modeling of diabetes using pluripotent stem cells. We discuss the current challenges and opportunities of these approaches to dissect ß-cell pathology and devise new pharmacological and cell replacement therapies. Stem Cells 2019;37:33-41.


Assuntos
Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Secretoras de Insulina/patologia , Animais , Diferenciação Celular , Humanos , Camundongos
13.
Elife ; 72018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30412052

RESUMO

Insulin gene mutations are a leading cause of neonatal diabetes. They can lead to proinsulin misfolding and its retention in endoplasmic reticulum (ER). This results in increased ER-stress suggested to trigger beta-cell apoptosis. In humans, the mechanisms underlying beta-cell failure remain unclear. Here we show that misfolded proinsulin impairs developing beta-cell proliferation without increasing apoptosis. We generated induced pluripotent stem cells (iPSCs) from people carrying insulin (INS) mutations, engineered isogenic CRISPR-Cas9 mutation-corrected lines and differentiated them to beta-like cells. Single-cell RNA-sequencing analysis showed increased ER-stress and reduced proliferation in INS-mutant beta-like cells compared with corrected controls. Upon transplantation into mice, INS-mutant grafts presented reduced insulin secretion and aggravated ER-stress. Cell size, mTORC1 signaling, and respiratory chain subunits expression were all reduced in INS-mutant beta-like cells, yet apoptosis was not increased at any stage. Our results demonstrate that neonatal diabetes-associated INS-mutations lead to defective beta-cell mass expansion, contributing to diabetes development.


Assuntos
Diabetes Mellitus/genética , Estresse do Retículo Endoplasmático/genética , Células-Tronco Pluripotentes Induzidas/química , Proinsulina/genética , Animais , Apoptose/genética , Sistemas CRISPR-Cas/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Diabetes Mellitus/patologia , Retículo Endoplasmático/genética , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Recém-Nascido , Células Secretoras de Insulina/química , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Mutação , Proinsulina/química , Dobramento de Proteína , Análise de Sequência de RNA , Transdução de Sinais , Análise de Célula Única
14.
Cell Rep ; 19(2): 281-294, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28402852

RESUMO

Activating germline mutations in STAT3 were recently identified as a cause of neonatal diabetes mellitus associated with beta-cell autoimmunity. We have investigated the effect of an activating mutation, STAT3K392R, on pancreatic development using induced pluripotent stem cells (iPSCs) derived from a patient with neonatal diabetes and pancreatic hypoplasia. Early pancreatic endoderm differentiated similarly from STAT3K392R and healthy-control cells, but in later stages, NEUROG3 expression was upregulated prematurely in STAT3K392R cells together with insulin (INS) and glucagon (GCG). RNA sequencing (RNA-seq) showed robust NEUROG3 downstream targets upregulation. STAT3 mutation correction with CRISPR/Cas9 reversed completely the disease phenotype. STAT3K392R-activating properties were not explained fully by altered DNA-binding affinity or increased phosphorylation. Instead, reporter assays demonstrated NEUROG3 promoter activation by STAT3 in pancreatic cells. Furthermore, proteomic and immunocytochemical analyses revealed increased nuclear translocation of STAT3K392R. Collectively, our results demonstrate that the STAT3K392R mutation causes premature endocrine differentiation through direct induction of NEUROG3 expression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Diferenciação Celular/genética , Diabetes Mellitus/genética , Proteínas do Tecido Nervoso/biossíntese , Fator de Transcrição STAT3/genética , Autoimunidade/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sistemas CRISPR-Cas , Linhagem Celular , Diabetes Mellitus/etiologia , Diabetes Mellitus/patologia , Regulação da Expressão Gênica no Desenvolvimento , Glucagon/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Mutação , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , Fator de Transcrição STAT3/biossíntese
15.
Dev Dyn ; 240(8): 1989-2001, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21761482

RESUMO

Classical cadherins are important cell adhesion molecules specifying and separating brain nuclei and developmental compartments. Cadherin-22 (Cdh22) belongs to type II subfamily of classical cadherins, and is expressed at the midbrain-hindbrain boundary during early embryogenesis. In Fgfr1 mutant mouse embryos, which have a disturbed midbrain-hindbrain border, Cdh22 is down-regulated. Here, we studied expression of Cdh22 in developing mouse brain in more detail and compared it to expression of related family members. This revealed both complementary and overlapping patterns of Cdh22, Cdh11, Cdh8, and Cdh6 expression in distinct regions of the forebrain and midbrain. We used a mutated allele of Cdh22 to study its function in brain development. Loss of Cdh22 caused reduced postnatal viability. Despite strong Cdh22 expression in the developing brain, we did not observe defects in compartmentalization or abnormalities in the midbrain and forebrain nuclei in Cdh22 mutants. This may be explained by functional redundancy between type II cadherins.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/embriologia , Encéfalo/metabolismo , Caderinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Animais , Caderinas/genética , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Extremidades/anatomia & histologia , Extremidades/embriologia , Genótipo , Masculino , Camundongos , Morfogênese/fisiologia , Mutação , Taxa de Sobrevida , Testículo/anatomia & histologia , Testículo/embriologia , Testículo/metabolismo
16.
Dev Biol ; 349(2): 270-82, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21074523

RESUMO

For the correct development of the central nervous system, the balance between self-renewing and differentiating divisions of the neuronal progenitors must be tightly regulated. To maintain their self-renewing identity, the progenitors need to retain both apical and basal interfaces. However, the identities of fate-determining signals which cells receive via these connections, and the exact mechanism of their action, are poorly understood. The conditional inactivation of Fibroblast growth factor (FGF) receptors 1 and 2 in the embryonic mouse midbrain-hindbrain area results in premature neuronal differentiation. Here, we aim to elucidate the connection between FGF signaling and neuronal progenitor maintenance. Our results reveal that the loss of FGF signaling leads to downregulation of Hes1 and upregulation of Ngn2, Dll1, and p57 in the ventricular zone (VZ) cells, and that this increased neurogenesis occurs cell-autonomously. Yet the cell cycle progression, apico-basal-polarity, cell-cell connections, and the positioning of mitotic spindle in the mutant VZ appear unaltered. Interestingly, FGF8-protein is highly concentrated in the basal lamina. Thus, FGFs may act through basal processes of neuronal progenitors to maintain their progenitor status. Indeed, midbrain neuronal progenitors deprived in vitro of FGFs switched from symmetrical proliferative towards symmetrical neurogenic divisions. We suggest that FGF signaling in the midbrain VZ is cell-autonomously required for the maintenance of symmetrical proliferative divisions via Hes1-mediated repression of neurogenic genes.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Mesencéfalo/embriologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Transdução de Sinais/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bromodesoxiuridina , Proliferação de Células , Cruzamentos Genéticos , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Neurogênese/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição HES-1
17.
PLoS One ; 5(6): e10881, 2010 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-20532162

RESUMO

beta-Catenin is a multifunctional protein involved in both signalling by secreted factors of Wnt family and regulation of the cellular architecture. We show that beta-catenin stabilization in mouse midbrain-rhombomere 1 region leads to robust up-regulation of several Wnt signalling target genes, including Fgf8. Suggestive of direct transcriptional regulation of the Fgf8 gene, beta-catenin stabilization resulted in Fgf8 up-regulation also in other tissues, specifically in the ventral limb ectoderm. Interestingly, stabilization of beta-catenin rapidly caused down-regulation of the expression of Wnt1 itself, suggesting a negative feedback loop. The changes in signal molecule expression were concomitant with deregulation of anterior-posterior and dorso-ventral patterning. The transcriptional regulatory functions of beta-catenin were confirmed by beta-catenin loss-of-function experiments. Temporally controlled inactivation of beta-catenin revealed a cell-autonomous role for beta-catenin in the maintenance of cell-type specific gene expression in the progenitors of midbrain dopaminergic neurons. These results highlight the role of beta-catenin in establishment of neuroectodermal signalling centers, promoting region-specific gene expression and regulation of cell fate determination.


Assuntos
Mesencéfalo/metabolismo , Transdução de Sinais/fisiologia , Transcrição Gênica/fisiologia , beta Catenina/fisiologia , Animais , Padronização Corporal , Fator 8 de Crescimento de Fibroblasto/genética , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Mesencéfalo/embriologia , Camundongos , Camundongos Transgênicos , Proteína Wnt1/genética
18.
PLoS One ; 4(2): e4341, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19194496

RESUMO

Conditional gene targeting strategies are progressively used to study gene function tissue-specifically and/or at a defined time period. Instrumental to all of these strategies is the generation of targeting vectors, and any methodology that would streamline the procedure would be highly beneficial. We describe a comprehensive transposition-based strategy to produce gene-targeting vectors for the generation of mouse conditional alleles. The system employs a universal cloning vector and two custom-designed mini-Mu transposons. It produces targeting constructions directly from BAC clones, and the alleles generated are modifiable by Cre and Flp recombinases. We demonstrate the applicability of the methodology by modifying two mouse genes, Chd22 and Drapc1. This straightforward strategy should be readily suitable for high-throughput targeting vector production.


Assuntos
DNA Nucleotidiltransferases/metabolismo , Elementos de DNA Transponíveis/genética , Marcação de Genes/métodos , Vetores Genéticos/genética , Integrases/metabolismo , Alelos , Animais , Sítios de Ligação Microbiológicos , Caderinas/genética , Células-Tronco Embrionárias/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Reprodutibilidade dos Testes
19.
J Neurosci ; 27(32): 8581-92, 2007 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-17687036

RESUMO

Fibroblast growth factors (FGFs) secreted from the midbrain-rhombomere 1 (r1) boundary instruct cell behavior in the surrounding neuroectoderm. For example, a combination of FGF and sonic hedgehog (SHH) can induce the development of the midbrain dopaminergic neurons, but the mechanisms behind the action and integration of these signals are unclear. We studied how FGF receptors (FGFRs) regulate cellular responses by analyzing midbrain-r1 development in mouse embryos, which carry different combinations of mutant Fgfr1, Fgfr2, and Fgfr3 alleles. Our results show that the FGFRs act redundantly to support cell survival in the dorsal neuroectoderm, promote r1 tissue identity, and regulate the production of ventral neuronal populations, including midbrain dopaminergic neurons. The compound Fgfr mutants have apparently normal WNT/SHH signaling and neurogenic gene expression in the ventral midbrain, but the number of proliferative neural progenitors is reduced as a result of precocious neuronal differentiation. Our results suggest a SoxB1 family member, Sox3, as a potential FGF-induced transcription factor promoting progenitor renewal. We propose a model for regulation of progenitor cell self-renewal and neuronal differentiation by combinatorial intercellular signals in the ventral midbrain.


Assuntos
Mesencéfalo/embriologia , Neurônios/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Rombencéfalo/embriologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Mesencéfalo/citologia , Mesencéfalo/fisiologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Gravidez , Rombencéfalo/citologia , Rombencéfalo/fisiologia , Células-Tronco/citologia
20.
Dev Biol ; 303(1): 231-43, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17150206

RESUMO

The mid-/hindbrain organizer (MHO) is characterized by the expression of a network of genes, which controls the patterning and development of the prospective midbrain and anterior hindbrain. One key molecule acting at the MHO is the fibroblast growth factor (Fgf) 8. Ectopic expression of Fgf8 induces genes that are normally expressed at the mid-/hindbrain boundary followed by the induction of midbrain and anterior hindbrain structures. Inactivation of the Fgf receptor (Fgfr) 1 gene, which was thought to be the primary transducer of the Fgf8 signal at the MHO, in the mid-/hindbrain region, leads to a deletion of dorsal structures of the mid-/hindbrain region, whereas ventral tissues are less severely affected. This suggests that other Fgfrs might be responsible for ventral mid-/hindbrain region development. Here we report the analysis of Fgfr2 conditional knockout mice, lacking the Fgfr2 in the mid-/hindbrain region and of Fgfr3 knockout mice with respect to the mid-/hindbrain region. In both homozygous mouse mutants, patterning of the mid-/hindbrain region is not altered, neuronal populations develop normal and are maintained into adulthood. This analysis shows that the Fgfr2 and the Fgfr3 on their own are dispensable for the development of the mid-/hindbrain region. We suggest functional redundancy of Fgf receptors in the mid-/hindbrain region.


Assuntos
Padronização Corporal/fisiologia , Mesencéfalo/embriologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Rombencéfalo/embriologia , Animais , Fator 8 de Crescimento de Fibroblasto/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA