Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Commun ; 15(1): 5567, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38956087

RESUMO

Diabetes involves the death or dysfunction of pancreatic ß-cells. Analysis of bulk sequencing from human samples and studies using in vitro and in vivo models suggest that endoplasmic reticulum and inflammatory signaling play an important role in diabetes progression. To better characterize cell type-specific stress response, we perform multiplexed single-cell RNA sequencing to define the transcriptional signature of primary human islet cells exposed to endoplasmic reticulum and inflammatory stress. Through comprehensive pair-wise analysis of stress responses across pancreatic endocrine and exocrine cell types, we define changes in gene expression for each cell type under different diabetes-associated stressors. We find that ß-, α-, and ductal cells have the greatest transcriptional response. We utilize stem cell-derived islets to study islet health through the candidate gene CIB1, which was upregulated under stress in primary human islets. Our findings provide insights into cell type-specific responses to diabetes-associated stress and establish a resource to identify targets for diabetes therapeutics.


Assuntos
Estresse do Retículo Endoplasmático , Células Secretoras de Insulina , Ilhotas Pancreáticas , Humanos , Estresse do Retículo Endoplasmático/genética , Ilhotas Pancreáticas/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Análise de Célula Única , Células Secretoras de Glucagon/metabolismo , Análise de Sequência de RNA , Transcriptoma , Estresse Fisiológico
2.
BMC Genomics ; 25(1): 105, 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38267908

RESUMO

Diabetes cell replacement therapy has the potential to be transformed by human pluripotent stem cell-derived ß cells (SC-ß cells). However, the precise identity of SC-ß cells in relationship to primary fetal and adult ß-cells remains unclear. Here, we used single-cell sequencing datasets to characterize the transcriptional identity of islets from in vitro differentiation, fetal islets, and adult islets. Our analysis revealed that SC-ß cells share a core ß-cell transcriptional identity with human adult and fetal ß-cells, however SC-ß cells possess a unique transcriptional profile characterized by the persistent expression and activation of progenitor and neural-biased gene networks. These networks are present in SC-ß cells, irrespective of the derivation protocol used. Notably, fetal ß-cells also exhibit this neural signature at the transcriptional level. Our findings offer insights into the transcriptional identity of SC-ß cells and underscore the need for further investigation of the role of neural transcriptional networks in their development.


Assuntos
Células-Tronco Pluripotentes , Adulto , Humanos , Diferenciação Celular/genética , Feto , Redes Reguladoras de Genes , Análise de Célula Única
4.
Nat Cell Biol ; 25(6): 904-916, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37188763

RESUMO

Insulin-producing ß cells created from human pluripotent stem cells have potential as a therapy for insulin-dependent diabetes, but human pluripotent stem cell-derived islets (SC-islets) still differ from their in vivo counterparts. To better understand the state of cell types within SC-islets and identify lineage specification deficiencies, we used single-nucleus multi-omic sequencing to analyse chromatin accessibility and transcriptional profiles of SC-islets and primary human islets. Here we provide an analysis that enabled the derivation of gene lists and activity for identifying each SC-islet cell type compared with primary islets. Within SC-islets, we found that the difference between ß cells and awry enterochromaffin-like cells is a gradient of cell states rather than a stark difference in identity. Furthermore, transplantation of SC-islets in vivo improved cellular identities overtime, while long-term in vitro culture did not. Collectively, our results highlight the importance of chromatin and transcriptional landscapes during islet cell specification and maturation.


Assuntos
Insulinas , Ilhotas Pancreáticas , Células-Tronco Pluripotentes , Humanos , Multiômica , Diferenciação Celular/genética , Células-Tronco Pluripotentes/metabolismo , Cromatina/genética , Cromatina/metabolismo , Insulinas/metabolismo
5.
JCI Insight ; 7(18)2022 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-36134655

RESUMO

Wolfram syndrome is a rare genetic disorder largely caused by pathogenic variants in the WFS1 gene and manifested by diabetes mellitus, optic nerve atrophy, and progressive neurodegeneration. Recent genetic and clinical findings have revealed Wolfram syndrome as a spectrum disorder. Therefore, a genotype-phenotype correlation analysis is needed for diagnosis and therapeutic development. Here, we focus on the WFS1 c.1672C>T, p.R558C variant, which is highly prevalent in the Ashkenazi Jewish population. Clinical investigation indicated that patients carrying the homozygous WFS1 c.1672C>T, p.R558C variant showed mild forms of Wolfram syndrome phenotypes. Expression of WFS1 p.R558C was more stable compared with the other known recessive pathogenic variants associated with Wolfram syndrome. Human induced pluripotent stem cell-derived (iPSC-derived) islets (SC-islets) homozygous for WFS1 c.1672C>T variant recapitulated genotype-related Wolfram syndrome phenotypes. Enhancing residual WFS1 function through a combination treatment of chemical chaperones mitigated detrimental effects caused by the WFS1 c.1672C>T, p.R558C variant and increased insulin secretion in SC-islets. Thus, the WFS1 c.1672C>T, p.R558C variant causes a mild form of Wolfram syndrome phenotypes, which can be remitted with a combination treatment of chemical chaperones. We demonstrate that our patient iPSC-derived disease model provides a valuable platform for further genotype-phenotype analysis and therapeutic development for Wolfram syndrome.


Assuntos
Células-Tronco Pluripotentes Induzidas , Atrofia Óptica , Síndrome de Wolfram , Homozigoto , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas de Membrana/genética , Atrofia Óptica/genética , Atrofia Óptica/patologia , Síndrome de Wolfram/diagnóstico , Síndrome de Wolfram/genética , Síndrome de Wolfram/patologia
6.
Nat Protoc ; 16(9): 4109-4143, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34349281

RESUMO

We detail a six-stage planar differentiation methodology for generating human pluripotent stem cell-derived pancreatic ß cells (SC-ß cells) that secrete high amounts of insulin in response to glucose stimulation. This protocol first induces definitive endoderm by treatment with Activin A and CHIR99021, then generates PDX1+/NKX6-1+ pancreatic progenitors through the timed application of keratinocyte growth factor, SANT1, TPPB, LDN193189 and retinoic acid. Endocrine induction and subsequent SC-ß-cell specification is achieved with a cocktail consisting of the cytoskeletal depolymerizing compound latrunculin A combined with XXI, T3, ALK5 inhibitor II, SANT1 and retinoic acid. The resulting SC-ß cells and other endocrine cell types can then be aggregated into islet-like clusters for analysis and transplantation. This differentiation methodology takes ~34 d to generate functional SC-ß cells, plus an additional 1-2 weeks for initial stem cell expansion and final cell assessment. This protocol builds upon a large body of previous work for generating ß-like cells. In this iteration, we have eliminated the need for 3D culture during endocrine induction, allowing for the generation of highly functional SC-ß cells to be done entirely on tissue culture polystyrene. This change simplifies the differentiation methodology, requiring only basic stem cell culture experience as well as familiarity with assessment techniques common in biology laboratories. In addition to expanding protocol accessibility and simplifying SC-ß-cell generation, we demonstrate that this planar methodology is amenable for differentiating SC-ß cells from a wide variety of cell lines from various sources, broadening its applicability.


Assuntos
Técnicas de Cultura de Células , Diferenciação Celular , Células Secretoras de Insulina , Células-Tronco Pluripotentes , Linhagem Celular , Humanos
8.
Cell Rep ; 32(8): 108067, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32846125

RESUMO

Human pluripotent stem cells differentiated to insulin-secreting ß cells (SC-ß cells) in islet organoids could provide an unlimited cell source for diabetes cell replacement therapy. However, current SC-ß cells generated in vitro are transcriptionally and functionally immature compared to native adult ß cells. Here, we use single-cell transcriptomic profiling to catalog changes that occur in transplanted SC-ß cells. We find that transplanted SC-ß cells exhibit drastic transcriptional changes and mature to more closely resemble adult ß cells. Insulin and IAPP protein secretions increase upon transplantation, along with expression of maturation genes lacking with differentiation in vitro, including INS, MAFA, CHGB, and G6PC2. Other differentiated cell types, such as SC-α and SC-enterochromaffin (SC-EC) cells, also exhibit large transcriptional changes. This study provides a comprehensive resource for understanding human islet cell maturation and provides important insights into maturation of SC-ß cells and other SC-islet cell types to enable future differentiation strategy improvements.


Assuntos
Linfócitos B/metabolismo , Perfilação da Expressão Gênica/métodos , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Humanos , Camundongos
9.
Cell Rep ; 31(8): 107687, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32460030

RESUMO

Generation of insulin-secreting ß cells in vitro is a promising approach for diabetes cell therapy. Human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) are differentiated to ß cells (SC-ß cells) and mature to undergo glucose-stimulated insulin secretion, but molecular regulation of this defining ß cell phenotype is unknown. Here, we show that maturation of SC-ß cells is regulated by the transcription factor SIX2. Knockdown (KD) or knockout (KO) of SIX2 in SC-ß cells drastically limits glucose-stimulated insulin secretion in both static and dynamic assays, along with the upstream processes of cytoplasmic calcium flux and mitochondrial respiration. Furthermore, SIX2 regulates the expression of genes associated with these key ß cell processes, and its expression is restricted to endocrine cells. Our results demonstrate that expression of SIX2 influences the generation of human SC-ß cells in vitro.


Assuntos
Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Diferenciação Celular , Humanos , Transdução de Sinais
10.
Sci Transl Med ; 12(540)2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32321868

RESUMO

Differentiation of insulin-producing pancreatic ß cells from induced pluripotent stem cells (iPSCs) derived from patients with diabetes promises to provide autologous cells for diabetes cell replacement therapy. However, current approaches produce patient iPSC-derived ß (SC-ß) cells with poor function in vitro and in vivo. Here, we used CRISPR-Cas9 to correct a diabetes-causing pathogenic variant in Wolfram syndrome 1 (WFS1) in iPSCs derived from a patient with Wolfram syndrome (WS). After differentiation to ß cells with our recent six-stage differentiation strategy, corrected WS SC-ß cells performed robust dynamic insulin secretion in vitro in response to glucose and reversed preexisting streptozocin-induced diabetes after transplantation into mice. Single-cell transcriptomics showed that corrected SC-ß cells displayed increased insulin and decreased expression of genes associated with endoplasmic reticulum stress. CRISPR-Cas9 correction of a diabetes-inducing gene variant thus allows for robust differentiation of autologous SC-ß cells that can reverse severe diabetes in an animal model.


Assuntos
Diabetes Mellitus , Células-Tronco Pluripotentes Induzidas , Células Secretoras de Insulina , Animais , Diferenciação Celular , Diabetes Mellitus/metabolismo , Diabetes Mellitus/terapia , Edição de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos
11.
Nat Biotechnol ; 38(4): 460-470, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32094658

RESUMO

Generation of pancreatic ß cells from human pluripotent stem cells (hPSCs) holds promise as a cell replacement therapy for diabetes. In this study, we establish a link between the state of the actin cytoskeleton and the expression of pancreatic transcription factors that drive pancreatic lineage specification. Bulk and single-cell RNA sequencing demonstrated that different degrees of actin polymerization biased cells toward various endodermal lineages and that conditions favoring a polymerized cytoskeleton strongly inhibited neurogenin 3-induced endocrine differentiation. Using latrunculin A to depolymerize the cytoskeleton during endocrine induction, we developed a two-dimensional differentiation protocol for generating human pluripotent stem-cell-derived ß (SC-ß) cells with improved in vitro and in vivo function. SC-ß cells differentiated from four hPSC lines exhibited first- and second-phase dynamic glucose-stimulated insulin secretion. Transplantation of islet-sized aggregates of these cells rapidly reversed severe preexisting diabetes in mice at a rate close to that of human islets and maintained normoglycemia for at least 9 months.


Assuntos
Engenharia Celular/métodos , Citoesqueleto/metabolismo , Células Secretoras de Insulina/citologia , Células-Tronco Pluripotentes/citologia , Actinas/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Diabetes Mellitus/terapia , Endoderma/citologia , Endoderma/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Pluripotentes/metabolismo , Tiazolidinas/farmacologia , Transativadores/metabolismo , Moduladores de Tubulina/farmacologia
12.
Curr Opin Biomed Eng ; 16: 27-33, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33738370

RESUMO

The islets of Langerhans are complex tissues composed of several cell types that secrete hormones. Loss or dysfunction of the insulin-producing ß cells leads to dysregulation of blood glucose levels, resulting in diabetes. A major goal in cellular engineering has been to generate ß cells from stem cells for use in cell-based therapies. However, the presence of other cell types within these islets can mask important details about ß cells when using population-level assays. Single-cell RNA sequencing have enabled transcriptional assessment of individual cells within mixed populations. These technologies allow for accurate assessment of specific cell types and subtypes of ß cells. Studies investigating different stages of ß cell maturity have led to several insights into understanding islet development and diabetes pathology. Here, we highlight the key findings from the use of single-cell RNA sequencing on stem cell-derived and primary human islet cells found in different maturation and diabetic states.

13.
Acta Biomater ; 97: 272-280, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31446050

RESUMO

Differentiation of stem cells into functional replacement cells and tissues is a major goal of the regenerative medicine field. However, one limitation has been organization of differentiated cells into multi-cellular, three-dimensional assemblies. The islets of Langerhans contain many endocrine and non-endocrine cell types, such as insulin-producing ß cells and endothelial cells. Despite the potential importance of endothelial cells to islet function, facilitating interactions between endothelial cells and islet endocrine cell types already differentiated from human embryonic stem cells has been difficult in vitro. We have developed a strategy of assembling human embryonic stem cell-derived islet cells with endothelial cells into three-dimensional aggregates on a hydrogel. The resulting islet organoids express ß cell and other endocrine markers and are functional, capable of undergoing glucose-stimulated insulin secretion. This assembly was not observed on traditional tissue culture plastic and in aggregates generated in suspension culture, highlighting how physical culture conditions greatly influence the interactions among these cell types. These results provide a platform for evaluating the effects of the islet tissue microenvironment on human embryonic stem cell-derived ß cells and other islet endocrine cells to develop tissue engineered islets. STATEMENT OF SIGNIFICANCE: Differentiation of insulin-producing cells and tissues from human pluripotent stem cells is being investigated for diabetes cell replacement therapies. Despite successes generating ß cells, the cell type responsible for glucose-stimulated insulin secretion within the islets of Langerhans found in the pancreas, successful assembly with other non-endocrine cell types, particularly endothelial cells, has been technically challenging. The present study provides a platform for the assembly of endothelial cells with SC-ß and other endocrine cells, producing islet organoids that are functional and express ß cell markers, that can be used to study the islet microenvironment and islet tissue engineering.


Assuntos
Células Imobilizadas/metabolismo , Células Endoteliais/metabolismo , Hidrogéis/química , Células Secretoras de Insulina/metabolismo , Organoides/metabolismo , Células-Tronco/metabolismo , Linhagem Celular , Células Imobilizadas/citologia , Células Endoteliais/citologia , Humanos , Células Secretoras de Insulina/citologia , Organoides/citologia , Células-Tronco/citologia
14.
Stem Cell Reports ; 12(2): 351-365, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30661993

RESUMO

Recent advances in human pluripotent stem cell (hPSC) differentiation protocols have generated insulin-producing cells resembling pancreatic ß cells. While these stem cell-derived ß (SC-ß) cells are capable of undergoing glucose-stimulated insulin secretion (GSIS), insulin secretion per cell remains low compared with islets and cells lack dynamic insulin release. Herein, we report a differentiation strategy focused on modulating transforming growth factor ß (TGF-ß) signaling, controlling cellular cluster size, and using an enriched serum-free media to generate SC-ß cells that express ß cell markers and undergo GSIS with first- and second-phase dynamic insulin secretion. Transplantation of these cells into mice greatly improves glucose tolerance. These results reveal that specific time frames for inhibiting and permitting TGF-ß signaling are required during SC-ß cell differentiation to achieve dynamic function. The capacity of these cells to undergo GSIS with dynamic insulin release makes them a promising cell source for diabetes cellular therapy.


Assuntos
Células-Tronco Embrionárias/fisiologia , Células Secretoras de Insulina/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Glucose/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Pâncreas/metabolismo , Pâncreas/fisiologia , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
15.
Annu Rev Biomed Eng ; 19: 57-84, 2017 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-28226216

RESUMO

The immune system is governed by an immensely complex network of cells and both intracellular and extracellular molecular factors. It must respond to an ever-growing number of biochemical and biophysical inputs by eliciting appropriate and specific responses in order to maintain homeostasis. But as with any complex system, a plethora of false positives and false negatives can occur to generate dysregulated responses. Dysregulated immune responses are essential components of diverse inflammation-driven pathologies, including cancer, heart disease, and autoimmune disorders. Nanoscale biomaterials (i.e., nanobiomaterials) have emerged as highly customizable platforms that can be engineered to interact with and direct immune responses, holding potential for the design of novel and targeted approaches to redirect or inhibit inflammation. Here, we present recent developments of nanobiomaterials that were rationally designed to target and modulate inflammatory cells and biochemical pathways for the treatment of immune dysregulation.


Assuntos
Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/prevenção & controle , Nanocápsulas/administração & dosagem , Nanoestruturas/administração & dosagem , Nanoestruturas/química , Animais , Humanos , Nanocápsulas/química , Resultado do Tratamento
16.
ACS Nano ; 10(12): 11290-11303, 2016 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-27935698

RESUMO

Atherosclerosis, a leading cause of heart disease, results from chronic vascular inflammation that is driven by diverse immune cell populations. Nanomaterials may function as powerful platforms for diagnostic imaging and controlled delivery of therapeutics to inflammatory cells in atherosclerosis, but efficacy is limited by nonspecific uptake by cells of the mononuclear phagocytes system (MPS). MPS cells located in the liver, spleen, blood, lymph nodes, and kidney remove from circulation the vast majority of intravenously administered nanomaterials regardless of surface functionalization or conjugation of targeting ligands. Here, we report that nanostructure morphology alone can be engineered for selective uptake by dendritic cells (DCs), which are critical mediators of atherosclerotic inflammation. Employing near-infrared fluorescence imaging and flow cytometry as a multimodal approach, we compared organ and cellular level biodistributions of micelles, vesicles (i.e., polymersomes), and filomicelles, all assembled from poly(ethylene glycol)-bl-poly(propylene sulfide) (PEG-bl-PPS) block copolymers with identical surface chemistries. While micelles and filomicelles were respectively found to associate with liver macrophages and blood-resident phagocytes, polymersomes were exceptionally efficient at targeting splenic DCs (up to 85% of plasmacytoid DCs) and demonstrated significantly lower uptake by other cells of the MPS. In a mouse model of atherosclerosis, polymersomes demonstrated superior specificity for DCs (p < 0.005) in atherosclerotic lesions. Furthermore, significant differences in polymersome cellular biodistributions were observed in atherosclerotic compared to naïve mice, including impaired targeting of phagocytes in lymph nodes. These results present avenues for immunotherapies in cardiovascular disease and demonstrate that nanostructure morphology can be tailored to enhance targeting specificity.


Assuntos
Aterosclerose/terapia , Células Dendríticas , Nanoestruturas , Animais , Inflamação , Ligantes , Camundongos , Micelas , Polímeros
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA