Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nature ; 567(7746): 43-48, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30760930

RESUMO

Cell-identity switches, in which terminally differentiated cells are converted into different cell types when stressed, represent a widespread regenerative strategy in animals, yet they are poorly documented in mammals. In mice, some glucagon-producing pancreatic α-cells and somatostatin-producing δ-cells become insulin-expressing cells after the ablation of insulin-secreting ß-cells, thus promoting diabetes recovery. Whether human islets also display this plasticity, especially in diabetic conditions, remains unknown. Here we show that islet non-ß-cells, namely α-cells and pancreatic polypeptide (PPY)-producing γ-cells, obtained from deceased non-diabetic or diabetic human donors, can be lineage-traced and reprogrammed by the transcription factors PDX1 and MAFA to produce and secrete insulin in response to glucose. When transplanted into diabetic mice, converted human α-cells reverse diabetes and continue to produce insulin even after six months. Notably, insulin-producing α-cells maintain expression of α-cell markers, as seen by deep transcriptomic and proteomic characterization. These observations provide conceptual evidence and a molecular framework for a mechanistic understanding of in situ cell plasticity as a treatment for diabetes and other degenerative diseases.


Assuntos
Diabetes Mellitus/patologia , Diabetes Mellitus/terapia , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/patologia , Animais , Biomarcadores/análise , Linhagem da Célula/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Diabetes Mellitus/imunologia , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Feminino , Glucagon/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Glucagon/transplante , Glucose/farmacologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Masculino , Camundongos , Especificidade de Órgãos/efeitos dos fármacos , Polipeptídeo Pancreático/metabolismo , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/efeitos dos fármacos , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Proteômica , Análise de Sequência de RNA , Transativadores/genética , Transativadores/metabolismo , Transcriptoma , Transdução Genética
2.
Bioinformatics ; 36(24): 5706-5708, 2021 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-33367748

RESUMO

SUMMARY: Highly multiplexed imaging technologies enable spatial profiling of dozens of biomarkers in situ. Here, we describe cytomapper, a computational tool written in R, that enables visualization of pixel- and cell-level information obtained by multiplexed imaging. To illustrate its utility, we analysed 100 images obtained by imaging mass cytometry from a cohort of type 1 diabetes patients. In addition, cytomapper includes a Shiny application that allows hierarchical gating of cells based on marker expression and visualization of selected cells in corresponding images. AVAILABILITY AND IMPLEMENTATION: The cytomapper package can be installed via https://www.bioconductor.org/packages/release/bioc/html/cytomapper.html. Code for analysis and further instructions can be found at https://github.com/BodenmillerGroup/cytomapper_publication. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

3.
Sci Data ; 9(1): 44, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35140234

RESUMO

Epithelial-mesenchymal transition (EMT) equips breast cancer cells for metastasis and treatment resistance. However, detection, inhibition, and elimination of EMT-undergoing cells is challenging due to the intrinsic heterogeneity of cancer cells and the phenotypic diversity of EMT programs. We comprehensively profiled EMT transition phenotypes in four non-cancerous human mammary epithelial cell lines using a flow cytometry surface marker screen, RNA sequencing, and mass cytometry. EMT was induced in the HMLE and MCF10A cell lines and in the HMLE-Twist-ER and HMLE-Snail-ER cell lines by prolonged exposure to TGFß1 or 4-hydroxytamoxifen, respectively. Each cell line exhibited a spectrum of EMT transition phenotypes, which we compared to the steady-state phenotypes of fifteen luminal, HER2-positive, and basal breast cancer cell lines. Our data provide multiparametric insights at single-cell level into the phenotypic diversity of EMT at different time points and in four human cellular models. These insights are valuable to better understand the complexity of EMT, to compare EMT transitions between the cellular models used here, and for the design of EMT time course experiments.


Assuntos
Neoplasias da Mama , Transição Epitelial-Mesenquimal , Transcriptoma , Neoplasias da Mama/genética , Linhagem Celular , Transição Epitelial-Mesenquimal/genética , Feminino , Perfilação da Expressão Gênica , Humanos
4.
Cell Metab ; 29(3): 755-768.e5, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30713109

RESUMO

Type 1 diabetes (T1D) results from the autoimmune destruction of insulin-producing ß cells. A comprehensive picture of the changes during T1D development is lacking due to limited sample availability, inability to sample longitudinally, and the paucity of technologies enabling comprehensive tissue profiling. Here, we analyzed 1,581 islets from 12 human donors, including eight with T1D, using imaging mass cytometry (IMC). IMC enabled simultaneous measurement of 35 biomarkers with single-cell and spatial resolution. We performed pseudotime analysis of islets through T1D progression from snapshot data to reconstruct the evolution of ß cell loss and insulitis. Our analyses revealed that ß cell destruction is preceded by a ß cell marker loss and by recruitment of cytotoxic and helper T cells. The approaches described herein demonstrate the value of IMC for improving our understanding of T1D pathogenesis, and our data lay the foundation for hypothesis generation and follow-on experiments.


Assuntos
Biomarcadores/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Citometria por Imagem/métodos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Pâncreas/metabolismo , Progressão da Doença , Humanos , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/patologia , Pâncreas/patologia
5.
Int J Biochem Cell Biol ; 88: 226-235, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28119131

RESUMO

In addition to ß-cells, pancreatic islets contain α- and δ-cells, which respectively produce glucagon and somatostatin. The reprogramming of these two endocrine cell types into insulin producers, as observed after a massive ß-cell ablation in mice, may help restoring a functional ß-cell mass in type 1 diabetes. Yet, the spontaneous α-to-ß and δ-to-ß conversion processes are relatively inefficient in adult animals and the underlying epigenetic mechanisms remain unclear. Several studies indicate that the conserved chromatin modifiers DNA methyltransferase 1 (Dnmt1) and Enhancer of zeste homolog 2 (Ezh2) are important for pancreas development and restrict islet cell plasticity. Here, to investigate the role of these two enzymes in α- and δ-cell development and fate maintenance, we genetically inactivated them in each of these two cell types. We found that loss of Dnmt1 does not enhance the conversion of α- or δ-cells toward a ß-like fate. In addition, while Dnmt1 was dispensable for the development of these two cell types, we noticed a gradual loss of α-, but not δ-cells in adult mice. Finally, we found that Ezh2 inactivation does not enhance α-cell plasticity, and, contrary to what is observed in ß-cells, does not impair α-cell proliferation. Our results indicate that both Dnmt1 and Ezh2 play distinct roles in the different islet cell types.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Células Secretoras de Glucagon/metabolismo , Homeostase , Células Secretoras de Somatostatina/metabolismo , Animais , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/deficiência , Proteína Potenciadora do Homólogo 2 de Zeste/deficiência , Ativação Enzimática , Células Secretoras de Glucagon/citologia , Camundongos , Células Secretoras de Somatostatina/citologia
6.
Nat Commun ; 8: 15285, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28585564

RESUMO

During asymmetric division, fate assignation in daughter cells is mediated by the partition of determinants from the mother. In the fly sensory organ precursor cell, Notch signalling partitions into the pIIa daughter. Notch and its ligand Delta are endocytosed into Sara endosomes in the mother cell and they are first targeted to the central spindle, where they get distributed asymmetrically to finally be dispatched to pIIa. While the processes of endosomal targeting and asymmetry are starting to be understood, the machineries implicated in the final dispatch to pIIa are unknown. We show that Sara binds the PP1c phosphatase and its regulator Sds22. Sara phosphorylation on three specific sites functions as a switch for the dispatch: if not phosphorylated, endosomes are targeted to the spindle and upon phosphorylation of Sara, endosomes detach from the spindle during pIIa targeting.


Assuntos
Divisão Celular Assimétrica , Proteínas de Drosophila/metabolismo , Endossomos/metabolismo , Fuso Acromático/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem da Célula , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Fosforilação , Ligação Proteica , Subunidades Proteicas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais
7.
Endocrinology ; 158(4): 696-701, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28323959

RESUMO

In August 2016, several leaders in glucagon biology gathered for the European Association for the Study of Diabetes Hagedorn Workshop in Oxford, England. A key point of discussion focused on the need for basal insulin to allow for the therapeutic benefit of glucagon blockade in the treatment of diabetes. Among the most enlightening experimental results presented were findings from studies in which glucagon receptor-deficient mice were administered streptozotocin to destroy pancreatic ß cells or had undergone diphtheria toxin-induced ß cell ablation. This article summarizes key features of the discussion as a consensus was reached. Agents that antagonize glucagon may be of great benefit for the treatment of diabetes; however, sufficient levels of basal insulin are required for their therapeutic efficacy.


Assuntos
Glicemia , Diabetes Mellitus/tratamento farmacológico , Glucagon/antagonistas & inibidores , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Humanos
8.
Cell Metab ; 25(3): 622-634, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28215845

RESUMO

Insulin-producing pancreatic ß cells in mice can slowly regenerate from glucagon-producing α cells in settings like ß cell loss, but the basis of this conversion is unknown. Moreover, it remains unclear if this intra-islet cell conversion is relevant to diseases like type 1 diabetes (T1D). We show that the α cell regulators Aristaless-related homeobox (Arx) and DNA methyltransferase 1 (Dnmt1) maintain α cell identity in mice. Within 3 months of Dnmt1 and Arx loss, lineage tracing and single-cell RNA sequencing revealed extensive α cell conversion into progeny resembling native ß cells. Physiological studies demonstrated that converted α cells acquire hallmark ß cell electrophysiology and show glucose-stimulated insulin secretion. In T1D patients, subsets of glucagon-expressing cells show loss of DNMT1 and ARX and produce insulin and other ß cell factors, suggesting that DNMT1 and ARX maintain α cell identity in humans. Our work reveals pathways regulated by Arx and Dnmt1 that are sufficient for achieving targeted generation of ß cells from adult pancreatic α cells.


Assuntos
Envelhecimento/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Células Secretoras de Glucagon/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Sinalização do Cálcio/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Criança , Pré-Escolar , DNA (Citosina-5-)-Metiltransferase 1 , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Feminino , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Glucagon/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Glucose/farmacologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Análise de Sequência de RNA , Análise de Célula Única , Adulto Jovem
9.
Elife ; 52016 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-27092792

RESUMO

Glucagon secretion dysregulation in diabetes fosters hyperglycemia. Recent studies report that mice lacking glucagon receptor (Gcgr(-/-)) do not develop diabetes following streptozotocin (STZ)-mediated ablation of insulin-producing ß-cells. Here, we show that diabetes prevention in STZ-treated Gcgr(-/-) animals requires remnant insulin action originating from spared residual ß-cells: these mice indeed became hyperglycemic after insulin receptor blockade. Accordingly, Gcgr(-/-) mice developed hyperglycemia after induction of a more complete, diphtheria toxin (DT)-induced ß-cell loss, a situation of near-absolute insulin deficiency similar to type 1 diabetes. In addition, glucagon deficiency did not impair the natural capacity of α-cells to reprogram into insulin production after extreme ß-cell loss. α-to-ß-cell conversion was improved in Gcgr(-/-) mice as a consequence of α-cell hyperplasia. Collectively, these results indicate that glucagon antagonism could i) be a useful adjuvant therapy in diabetes only when residual insulin action persists, and ii) help devising future ß-cell regeneration therapies relying upon α-cell reprogramming.


Assuntos
Fármacos Gastrointestinais/metabolismo , Glucagon/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Transdução de Sinais , Animais , Diabetes Mellitus Experimental/fisiopatologia , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Receptores de Glucagon/deficiência
10.
Diabetes ; 60(11): 2872-82, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21926270

RESUMO

OBJECTIVE: To evaluate whether healthy or diabetic adult mice can tolerate an extreme loss of pancreatic α-cells and how this sudden massive depletion affects ß-cell function and blood glucose homeostasis. RESEARCH DESIGN AND METHODS: We generated a new transgenic model allowing near-total α-cell removal specifically in adult mice. Massive α-cell ablation was triggered in normally grown and healthy adult animals upon diphtheria toxin (DT) administration. The metabolic status of these mice was assessed in 1) physiologic conditions, 2) a situation requiring glucagon action, and 3) after ß-cell loss. RESULTS: Adult transgenic mice enduring extreme (98%) α-cell removal remained healthy and did not display major defects in insulin counter-regulatory response. We observed that 2% of the normal α-cell mass produced enough glucagon to ensure near-normal glucagonemia. ß-Cell function and blood glucose homeostasis remained unaltered after α-cell loss, indicating that direct local intraislet signaling between α- and ß-cells is dispensable. Escaping α-cells increased their glucagon content during subsequent months, but there was no significant α-cell regeneration. Near-total α-cell ablation did not prevent hyperglycemia in mice having also undergone massive ß-cell loss, indicating that a minimal amount of α-cells can still guarantee normal glucagon signaling in diabetic conditions. CONCLUSIONS: An extremely low amount of α-cells is sufficient to prevent a major counter-regulatory deregulation, both under physiologic and diabetic conditions. We previously reported that α-cells reprogram to insulin production after extreme ß-cell loss and now conjecture that the low α-cell requirement could be exploited in future diabetic therapies aimed at regenerating ß-cells by reprogramming adult α-cells.


Assuntos
Apoptose/efeitos dos fármacos , Células Secretoras de Glucagon/efeitos dos fármacos , Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores de Glucagon/metabolismo , Transdução de Sinais , Animais , Contagem de Células , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Toxina Diftérica/toxicidade , Glucagon/sangue , Glucagon/genética , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Hiperglicemia/induzido quimicamente , Hiperglicemia/prevenção & controle , Hipoglicemia/prevenção & controle , Insulina/sangue , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Pâncreas/patologia , Regiões Promotoras Genéticas , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Estreptozocina/toxicidade , Tamoxifeno/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA