Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 424
Filtrar
1.
Endocrinol Metab (Seoul) ; 39(2): 191-205, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38572534

RESUMO

In the quest to combat insulin-dependent diabetes mellitus (IDDM), allogenic pancreatic islet cell therapy sourced from deceased donors represents a significant therapeutic advance. However, the applicability of this approach is hampered by donor scarcity and the demand for sustained immunosuppression. Human induced pluripotent stem cells are a game-changing resource for generating synthetic functional insulin-producing ß cells. In addition, novel methodologies allow the direct expansion of pancreatic progenitors and mature ß cells, thereby circumventing prolonged differentiation. Nevertheless, achieving practical reproducibility and scalability presents a substantial challenge for this technology. As these innovative approaches become more prominent, it is crucial to thoroughly evaluate existing expansion techniques with an emphasis on their optimization and scalability. This manuscript delineates these cutting-edge advancements, offers a critical analysis of the prevailing strategies, and underscores pivotal challenges, including cost-efficiency and logistical issues. Our insights provide a roadmap, elucidating both the promises and the imperatives in harnessing the potential of these cellular therapies for IDDM.


Assuntos
Diabetes Mellitus Tipo 1 , Células-Tronco Pluripotentes Induzidas , Células Secretoras de Insulina , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/transplante , Células-Tronco Pluripotentes Induzidas/citologia , Diabetes Mellitus Tipo 1/terapia , Transplante das Ilhotas Pancreáticas/métodos , Diferenciação Celular , Insulina/metabolismo , Animais , Técnicas de Cultura de Células/métodos
2.
J Colloid Interface Sci ; 636: 90-102, 2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-36623370

RESUMO

Type 1 diabetes mellitus is an auto-immune disease causing the T-cell mediated destruction of insulin-producing ß-cells, resulting in chronic hyperglycemia. Current treatments such as insulin replacement therapy or the transplantation of pancreas or pancreatic islets present major disadvantages such as the constant need of drugs, as well as a shortage of donor organs. In this review, we discuss a sustainable solution to overcome these limitations combining the use of ß-cells, derived from stem cells, and their encapsulation within a protective matrix. This article provides an exhaustive overview of currently investigated stem cell sources including embryonic, mesenchymal as well as induced pluripotent stem cells in combination with various up to date encapsulation methods allowing the formation of immuno-protective devices. In order to identify current limitations of this interdisciplinary therapeutic approach and to find sustainable solutions, it is essential to consider key aspects from all involved domains. This includes biological parameters such as the stem cell origin but also the different aspects of the encapsulation process, the used materials and their physico-chemical properties such as elasticity, porosity and permeability cut-off as well as the best implantation sites allowing efficient and self-autonomous control of glycemia by the transplanted encapsulated cells.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Humanos , Diabetes Mellitus Tipo 1/terapia , Células-Tronco , Pâncreas , Células Secretoras de Insulina/transplante , Insulina , Diferenciação Celular
3.
Cell Transplant ; 31: 9636897221096160, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35583214

RESUMO

Patients fulfilling criteria for euthanasia can choose to donate their organs after circulatory death [donors after euthanasia (DCD V)]. This study assesses the outcome of islet cell isolation from DCD V pancreases. A procedure for DCD V procurement provided 13 pancreases preserved in Institut Georges Lopez-1 preservation solution and following acirculatory warm ischemia time under 10 minutes. Islet cell isolation outcomes are compared with those from reference donors after brain death (DBD, n = 234) and a cohort of donors after controlled circulatory death (DCD III, n = 29) procured under the same conditions. Islet cell isolation from DCD V organs resulted in better in vitro outcome than for selected DCD III or reference DBD organs. A 50% higher average beta cell number before and after culture and a higher average beta cell purity (35% vs 24% and 25%) was observed, which led to more frequent selection for our clinical protocol (77% of isolates vs 50%). The functional capacity of a DCD V islet cell preparation was illustrated by its in vivo effect following intraportal transplantation in a type 1 diabetes patient: injection of 2 million beta cells/kg body weight (1,900 IEQ/kg body weight) at 39% insulin purity resulted in an implant with functional beta cell mass that represented 30% of that in non-diabetic controls. In conclusion, this study describes procurement and preservation conditions for donor organs after euthanasia, which allow preparation of cultured islet cells, that more frequently meet criteria for clinical use than those from DBD or DCD III organs.


Assuntos
Células Secretoras de Insulina , Doadores de Tecidos , Peso Corporal , Morte Encefálica , Eutanásia , Humanos , Células Secretoras de Insulina/transplante , Pâncreas
4.
Sci Rep ; 12(1): 5852, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35393479

RESUMO

Insulin-producing cells (IPCs) generated by our established protocol have reached the non-clinical 'proof of concept' stage. Our strategy for their clinical application is the autotransplantation of IPCs into patients with type 1 diabetes mellitus (T1DM). In this context, the autoimmunity that characterized T1DM is important, rather than allorejection. We aimed to determine how these IPCs respond to T1DM autoimmunity. IPCs were generated from the subcutaneous fat tissue of non-obese diabetic (NOD) mice using our protocol. IPCs derived from NOD mice were transplanted under the kidney capsules of NOD mice at the onset of diabetes and the subsequent changes in blood glucose concentration were characterized. Blood glucose decreased within 30 days of transplantation, but increased again after 40-60 days in three of four recipient NOD mice. In tissue samples, the numbers of CD4+ and CD8+ T cells were significantly higher 60 days after transplantation than 30 days after transplantation. In conclusion, IPCs significantly ameliorate the diabetes of mice in the short term, but are damaged by autoimmunity in the longer term, as evidenced by local T cells accumulation. This study provides new insights into potential stem cell therapies for T1DM.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Animais , Autoimunidade , Glicemia , Linfócitos T CD8-Positivos , Diabetes Mellitus Tipo 1/terapia , Humanos , Insulina , Células Secretoras de Insulina/transplante , Camundongos , Camundongos Endogâmicos NOD , Células-Tronco
5.
Front Endocrinol (Lausanne) ; 12: 734360, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34912295

RESUMO

Diabetic retinopathy (DR) is a potentially devastating complication of diabetes because it puts patients at risk of blindness. Diabetes is a common cause of blindness in the U.S. and worldwide and is dramatically increasing in global prevalence. Thus new approaches are needed to prevent this dreaded complication. There is extensive data that indicates beta cell secretory failure is a risk factor for DR, independent of its influence on glycemic control. This perspective article will provide evidence for insufficient endogenous insulin secretion as an important factor in the development of DR. The areas of evidence discussed are: (a) Presence of insulin receptors in the retina, (b) Clinical studies that show an association of beta cell insufficiency with DR, (c) Treatment with insulin in type 2 diabetes, a marker for endogenous insulin deficiency, is an independent risk factor for DR, (d) Recent clinical studies that link DR with an insulin deficient form of type 2 diabetes, and (e) Beta cell replacement studies that demonstrate endogenous insulin prevents progression of DR. The cumulative data drive our conclusion that beta cell replacement will have an important role in preventing DR and/or mitigating its severity in both type 1 diabetes and insulinopenic type 2 diabetes.


Assuntos
Cegueira/prevenção & controle , Retinopatia Diabética/metabolismo , Retinopatia Diabética/terapia , Células Secretoras de Insulina/transplante , Insulina/deficiência , Cegueira/etiologia , Estudos Clínicos como Assunto , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Retinopatia Diabética/etiologia , Humanos , Insulina/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Fatores de Risco
6.
Lancet Diabetes Endocrinol ; 9(10): 708-724, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34480875

RESUMO

Type 1 diabetes is characterised by autoimmune-mediated destruction of pancreatic ß-cell mass. With the advent of insulin therapy a century ago, type 1 diabetes changed from a progressive, fatal disease to one that requires lifelong complex self-management. Replacing the lost ß-cell mass through transplantation has proven successful, but limited donor supply and need for lifelong immunosuppression restricts widespread use. In this Review, we highlight incremental advances over the past 20 years and remaining challenges in regenerative medicine approaches to restoring ß-cell mass and function in type 1 diabetes. We begin by summarising the role of endocrine islets in glucose homoeostasis and how this is altered in disease. We then discuss the potential regenerative capacity of the remaining islet cells and the utility of stem cell-derived ß-like cells to restore ß-cell function. We conclude with tissue engineering approaches that might improve the engraftment, function, and survival of ß-cell replacement therapies.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Ilhotas Pancreáticas/fisiologia , Medicina Regenerativa , Animais , Contagem de Células , Proliferação de Células/fisiologia , Diabetes Mellitus Tipo 1/fisiopatologia , História do Século XXI , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/história , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/tendências , Regeneração/fisiologia , Medicina Regenerativa/história , Medicina Regenerativa/métodos , Medicina Regenerativa/tendências , Engenharia Tecidual/história , Engenharia Tecidual/métodos , Engenharia Tecidual/tendências
7.
STAR Protoc ; 2(3): 100675, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34345868

RESUMO

Stem cell-derived beta cells (SC-ß-cells) engrafted into mice serve as a pre-clinical model of diabetes. It is helpful to recover viable ß cells following transplantation to perform tests on the graft. We developed a protocol to retrieve and purify a sufficient number of live ß cells from mice following long-term human SC-ß-cell engraftment. The protocol enables examination of SC-ß-cells undergoing developmental and metabolic changes in vivo and may facilitate the understanding of metabolic demand on SC-ß-cells.


Assuntos
Separação Celular/métodos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/transplante , Animais , Xenoenxertos , Humanos , Transplante das Ilhotas Pancreáticas/métodos , Camundongos , Células-Tronco/citologia
8.
Science ; 373(6554): 516-522, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34326233

RESUMO

Technological advancements in blood glucose monitoring and therapeutic insulin administration have improved the quality of life for people with type 1 diabetes. However, these efforts fall short of replicating the exquisite metabolic control provided by native islets. We examine the integrated advancements in islet cell replacement and immunomodulatory therapies that are coalescing to enable the restoration of endogenous glucose regulation. We highlight advances in stem cell biology and graft site design, which offer innovative sources of cellular material and improved engraftment. We also cover cutting-edge approaches for preventing allograft rejection and recurrent autoimmunity. These insights reflect a growing understanding of type 1 diabetes etiology, ß cell biology, and biomaterial design, together highlighting therapeutic opportunities to durably replace the ß cells destroyed in type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Imunomodulação , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas , Animais , Autoimunidade , Glicemia/metabolismo , Diferenciação Celular , Engenharia Celular , Microambiente Celular , Diabetes Mellitus Tipo 1/metabolismo , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Humanos , Tolerância Imunológica , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Células-Tronco Pluripotentes/transplante , Transplante de Células-Tronco
9.
In Vitro Cell Dev Biol Anim ; 57(6): 587-597, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34212340

RESUMO

Conventional methods for obtaining pancreatic ß cells are based on simulating the embryonic development phase of endocrine cells via hierarchical differentiation of pluripotent stem cells (PSCs). Accordingly, we attempted to modify the protocols for obtaining insulin-secreting cells (ISCs) by sequential differentiation of a human embryonic stem cell (hESC), using the HS181 cell line. Furthermore, we hypothesize that actual pancreatic endocrine cells may arise from trans-differentiation of mature ductal cells after the embryonic developmental stage and throughout the rest of life. According to the hypothesis, ductal cells are trans-differentiated into endocrine and exocrine cells, undergoing a partial epithelial to mesenchymal transition (EMT). To address this issue, we developed two new protocols based on hESC differentiation to obtain ductal cells and then induce EMT in cells to obtain hormone-secreting islet-like cells (HSCs). The ductal (pre-EMT exocrine) cells were then induced to undergo partial EMT by treating with Wnt3a and activin A, in hypoxia. The cell derived from the latter method significantly expressed the main endocrine cell-specific markers and also ß cells, in particular. These experiments not only support our hypothetical model but also offer a promising approach to develop new methods to compensate ß cell depletion in patients with type 1 diabetes mellitus (T1DM). Although this protocol of generating islet-like cells from ductal cells has a potential to treat T1DM, this strategy may be exploited to optimize the function of these cells in an animal model and future clinical applications.


Assuntos
Transdiferenciação Celular/genética , Diabetes Mellitus Tipo 1/terapia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Pluripotentes/citologia , Proteína Wnt3A/genética , Técnicas de Cultura de Células , Diferenciação Celular/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Células Endócrinas/citologia , Transição Epitelial-Mesenquimal/genética , Células-Tronco Embrionárias Humanas/transplante , Humanos , Insulina/genética , Insulina/metabolismo , Secreção de Insulina/genética , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/transplante , Pâncreas/crescimento & desenvolvimento , Pâncreas/patologia , Células-Tronco Pluripotentes/transplante
11.
Adv Sci (Weinh) ; 8(17): e2003708, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34258870

RESUMO

Islet transplantation has shown promise as a curative therapy for type 1 diabetes (T1D). However, the side effects of systemic immunosuppression and limited long-term viability of engrafted islets, together with the scarcity of donor organs, highlight an urgent need for the development of new, improved, and safer cell-replacement strategies. Induction of local immunotolerance to prevent allo-rejection against islets and stem cell derived ß cells has the potential to improve graft function and broaden the applicability of cellular therapy while minimizing adverse effects of systemic immunosuppression. In this mini review, recent developments in non-encapsulation, local immunomodulatory approaches for T1D cell replacement therapies, including islet/ß cell modification, immunomodulatory biomaterial platforms, and co-transplantation of immunomodulatory cells are discussed. Key advantages and remaining challenges in translating such technologies to clinical settings are identified. Although many of the studies discussed are preliminary, the growing interest in the field has led to the exploration of new combinatorial strategies involving cellular engineering, immunotherapy, and novel biomaterials. Such interdisciplinary research will undoubtedly accelerate the development of therapies that can benefit the whole T1D population.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/cirurgia , Rejeição de Enxerto/prevenção & controle , Imunomodulação/imunologia , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas/métodos , Rejeição de Enxerto/imunologia , Humanos
12.
Methods Mol Biol ; 2312: 35-57, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34228283

RESUMO

Diabetes mellitus is a complex metabolic disease characterized by chronically deregulated blood-glucose levels. To restore glucose homeostasis, therapeutic strategies allowing well-controlled production and release of insulinogenic hormones into the blood circulation are required. In this chapter, we describe how mammalian cells can be engineered for applications in diabetes treatment. While closed-loop control systems provide automated and self-sufficient synchronization of glucose sensing and drug production, drug production in open-loop control systems is engineered to depend on exogenous user-defined trigger signals. Rational, robust, and reliable manufacture practices for mammalian cell engineering are essential for industrial-scale mass-production in view of clinical and commercial applications.


Assuntos
Técnicas Biossensoriais , Glicemia/metabolismo , Engenharia Celular , Diabetes Mellitus Experimental/terapia , Terapia Genética , Peptídeo 1 Semelhante ao Glucagon/genética , Células Secretoras de Insulina/transplante , Insulina/genética , Biologia Sintética , Animais , Biomarcadores/sangue , Encapsulamento de Células , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/diagnóstico , Diabetes Mellitus Experimental/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Células HEK293 , Homeostase , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Transfecção
13.
Front Immunol ; 12: 690623, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34248981

RESUMO

Mesenchymal stem cell (MSC)-based therapy for type 1 diabetes mellitus (T1DM) has been the subject matter of many studies over the past few decades. The wide availability, negligible teratogenic risks and differentiation potential of MSCs promise a therapeutic alternative to traditional exogenous insulin injections or pancreatic transplantation. However, conflicting arguments have been reported regarding the immunological profile of MSCs. While some studies support their immune-privileged, immunomodulatory status and successful use in the treatment of several immune-mediated diseases, others maintain that allogeneic MSCs trigger immune responses, especially following differentiation or in vivo transplantation. In this review, the intricate mechanisms by which MSCs exert their immunomodulatory functions and the influencing variables are critically addressed. Furthermore, proposed avenues to enhance these effects, including cytokine pretreatment, coadministration of mTOR inhibitors, the use of Tregs and gene manipulation, are presented. As an alternative, the selection of high-benefit, low-risk donors based on HLA matching, PD-L1 expression and the absence of donor-specific antibodies (DSAs) are also discussed. Finally, the necessity for the transplantation of human MSC (hMSC)-derived insulin-producing cells (IPCs) into humanized mice is highlighted since this strategy may provide further insights into future clinical applications.


Assuntos
Glicemia/metabolismo , Diferenciação Celular , Diabetes Mellitus Tipo 1/cirurgia , Células Secretoras de Insulina/transplante , Insulina/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/imunologia , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Células-Tronco Mesenquimais/imunologia , Fenótipo
15.
J Clin Endocrinol Metab ; 106(10): 3049-3059, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34061967

RESUMO

CONTEXT: The Igls criteria were developed to provide a consensus definition for outcomes of ß-cell replacement therapy in the treatment of diabetes during a January 2017 workshop sponsored by the International Pancreas & Islet Transplant Association (IPITA) and the European Pancreas & Islet Transplant Association. In July 2019, a symposium at the 17th IPITA World Congress was held to examine the Igls criteria after 2 years in clinical practice, including validation against continuous glucose monitoring (CGM)-derived glucose targets, and to propose future refinements that would allow for comparison of outcomes with artificial pancreas system approaches. EVIDENCE ACQUISITION: Utilization of the criteria in various clinical and research settings was illustrated by population as well as individual outcome data of 4 islet and/or pancreas transplant centers. Validation against CGM metrics was conducted in 55 islet transplant recipients followed-up to 10 years from a fifth center. EVIDENCE SYNTHESIS: The Igls criteria provided meaningful clinical assessment on an individual patient and treatment group level, allowing for comparison both within and between different ß-cell replacement modalities. Important limitations include the need to account for changes in insulin requirements and C-peptide levels relative to baseline. In islet transplant recipients, CGM glucose time in range improved with each category of increasing ß-cell graft function. CONCLUSIONS: Future Igls 2.0 criteria should consider absolute rather than relative levels of insulin use and C-peptide as qualifiers with treatment success based on glucose assessment using CGM metrics on par with assessment of glycated hemoglobin and severe hypoglycemia events.


Assuntos
Automonitorização da Glicemia/normas , Diabetes Mellitus/terapia , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas/normas , Avaliação de Resultados em Cuidados de Saúde/normas , Glicemia/análise , Hemoglobinas Glicadas/análise , Humanos , Resultado do Tratamento
16.
J Diabetes Investig ; 12(9): 1542-1544, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34110690

RESUMO

Three complementary approaches for type 1 diabetes. Immunotherapy targets the pathogenic immune cells or inflammatory cytokines to revert type 1 diabetes. An artificial pancreas delivers insulin automatically using continuous glucose monitoring, a controlling algorithm, and an insulin pump. Beta cell replacement therapy varies depending on the cell sources: allogeneic, or xenogeneic islet; beta-like cells derived from ESCs or iPSCs.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Imunoterapia/métodos , Sistemas de Infusão de Insulina/estatística & dados numéricos , Células Secretoras de Insulina/transplante , Pâncreas Artificial/estatística & dados numéricos , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Humanos , Prognóstico
17.
Cell Metab ; 33(4): 721-731, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33826915

RESUMO

Tremendous progress has been made over the last two decades in the field of pancreatic beta cell replacement therapy as a curative measure for diabetes. Transplantation studies have demonstrated therapeutic efficacy, and cGMP-grade cell products are currently being deployed for the first time in human clinical trials. In this perspective, we discuss current challenges surrounding the generation, delivery, and engraftment of stem cell-derived islet-like cells, along with strategies to induce durable tolerance to grafted cells, with an eye toward a functional cellular-based therapy enabling insulin independence for patients with diabetes.


Assuntos
Insulina/metabolismo , Medicina Regenerativa , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/terapia , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
Cells ; 10(1)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33477961

RESUMO

Diabetes mellitus is a pandemic metabolic disorder that results from either the autoimmune destruction or the dysfunction of insulin-producing pancreatic beta cells. A promising cure is beta cell replacement through the transplantation of islets of Langerhans. However, donor shortage hinders the widespread implementation of this therapy. Human pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells, represent an attractive alternative beta cell source for transplantation. Although major advances over the past two decades have led to the generation of stem cell-derived beta-like cells that share many features with genuine beta cells, producing fully mature beta cells remains challenging. Here, we review the current status of beta cell differentiation protocols and highlight specific challenges that are associated with producing mature beta cells. We address the challenges and opportunities that are offered by monogenic forms of diabetes. Finally, we discuss the remaining hurdles for clinical application of stem cell-derived beta cells and the status of ongoing clinical trials.


Assuntos
Diferenciação Celular , Diabetes Mellitus Tipo 1 , Células-Tronco Embrionárias Humanas , Células-Tronco Pluripotentes Induzidas , Células Secretoras de Insulina , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/terapia , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/patologia , Células-Tronco Embrionárias Humanas/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/transplante , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/transplante
19.
Stem Cells ; 39(5): 522-535, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33497522

RESUMO

Strategies to mitigate the pathologies from diabetes range from simply administering insulin to prescribing complex drug/biologic regimens combined with lifestyle changes. There is a substantial effort to better understand ß-cell physiology during diabetes pathogenesis as a means to develop improved therapies. The convergence of multiple fields ranging from developmental biology to microfluidic engineering has led to the development of new experimental systems to better study complex aspects of diabetes and ß-cell biology. Here we discuss the available insulin-secreting cell types used in research, ranging from primary human ß-cells, to cell lines, to pluripotent stem cell-derived ß-like cells. Each of these sources possess inherent strengths and weaknesses pertinent to specific applications, especially in the context of engineered platforms. We then outline how insulin-expressing cells have been used in engineered platforms and how recent advances allow for better mimicry of in vivo conditions. Chief among these conditions are ß-cell interactions with other endocrine organs. This facet is beginning to be thoroughly addressed by the organ-on-a-chip community, but holds enormous potential in the development of novel diabetes therapeutics. Furthermore, high throughput strategies focused on studying ß-cell biology, improving ß-cell differentiation, or proliferation have led to enormous contributions in the field and will no doubt be instrumental in bringing new diabetes therapeutics to the clinic.


Assuntos
Diabetes Mellitus/terapia , Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , Células-Tronco Pluripotentes/metabolismo , Comunicação Celular/genética , Diferenciação Celular/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Humanos , Insulina/genética , Células Secretoras de Insulina/transplante , Dispositivos Lab-On-A-Chip , Células-Tronco Pluripotentes/transplante
20.
Biochem Biophys Res Commun ; 534: 1053-1058, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33160622

RESUMO

High mobility group (HMGB1) is an alarmin known to be harmful to pancreatic beta cells and associated with diabetes mellitus pathogenesis and pancreatic islet graft failure. It has been long thought that the suppression of HMGB1 molecule is beneficial to the beta cells. However, recent studies have indicated that cytoplasmic HMGB1 (cHMGB1) could function as a modulator to relieve cells from apoptotic stress by autophagy induction. Particularly, pancreatic beta cells have been known to utilize the autophagy-to-apoptosis switch when exposed to hypoxia or lipotoxicity. This study aimed to investigate the beta cells under hypoxic and lipotoxic stress while utilizing a small molecule inhibitor of HMGB1, inflachromene (ICM) which can suppress cHMGB1 accumulation. It was revealed that under cellular stress, blockade of cHMGB1 accumulation decreased the viability of islet grafts, primary islets and MIN6 cells. MIN6 cells under cHMGB1 blockade along with lipotoxic stress showed decreased autophagic flux and increased apoptosis. Moreover, cHMGB1 blockade in HFD-fed mice produced unfavorable outcomes on their glucose tolerance. In sum, these results suggested the role of cHMGB1 within beta cell autophagy/apoptosis checkpoint. Given the importance of autophagy in beta cells under apoptotic stresses, this study might provide further insights regarding HMGB1 and diabetes.


Assuntos
Proteína HMGB1/antagonistas & inibidores , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Proteína HMGB1/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...