Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Diabetologia ; 67(9): 1912-1929, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38871836

RESUMO

AIMS/HYPOTHESIS: Stem cell-derived islets (SC-islets) are being used as cell replacement therapy for insulin-dependent diabetes. Non-invasive long-term monitoring methods for SC-islet grafts, which are needed to detect misguided differentiation in vivo and to optimise their therapeutic effectiveness, are lacking. Positron emission tomography (PET) has been used to monitor transplanted primary islets. We therefore aimed to apply PET as a non-invasive monitoring method for SC-islet grafts. METHODS: We implanted different doses of human SC-islets, SC-islets derived using an older protocol or a state-of-the-art protocol and SC-islets genetically rendered hyper- or hypoactive into mouse calf muscle to yield different kinds of grafts. We followed the grafts with PET using two tracers, glucagon-like peptide 1 receptor-binding [18F]F-dibenzocyclooctyne-exendin-4 ([18F]exendin) and the dopamine precursor 6-[18F]fluoro-L-3,4-dihydroxyphenylalanine ([18F]FDOPA), for 5 months, followed by histological assessment of graft size and composition. Additionally, we implanted a kidney subcapsular cohort with different SC-islet doses to assess the connection between C-peptide and stem cell-derived beta cell (SC-beta cell) mass. RESULTS: Small but pure and large but impure grafts were derived from SC-islets. PET imaging allowed detection of SC-islet grafts even <1 mm3 in size, [18F]exendin having a better detection rate than [18F]FDOPA (69% vs 44%, <1 mm3; 96% vs 85%, >1 mm3). Graft volume quantified with [18F]exendin (r2=0.91) and [18F]FDOPA (r2=0.86) strongly correlated with actual graft volume. [18F]exendin PET delineated large cystic structures and its uptake correlated with graft SC-beta cell proportion (r2=0.68). The performance of neither tracer was affected by SC-islet graft hyper- or hypoactivity. C-peptide measurements under fasted or glucose-stimulated conditions did not correlate with SC-islet graft volume or SC-beta cell mass, with C-peptide under hypoglycaemia having a weak correlation with SC-beta cell mass (r2=0.52). CONCLUSIONS/INTERPRETATION: [18F]exendin and [18F]FDOPA PET enable non-invasive assessment of SC-islet graft size and aspects of graft composition. These methods could be leveraged for optimising SC-islet cell replacement therapy in diabetes.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Tomografia por Emissão de Pósitrons , Transplante das Ilhotas Pancreáticas/métodos , Animais , Camundongos , Humanos , Tomografia por Emissão de Pósitrons/métodos , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Masculino , Diabetes Mellitus Tipo 1/cirurgia , Diabetes Mellitus Tipo 1/metabolismo , Feminino
2.
Biochem Biophys Res Commun ; 700: 149509, 2024 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-38306929

RESUMO

Optic neuropathies, such as glaucoma, are due to progressive retinal ganglion cells (RGCs) degeneration, result in irreversible vision loss. The promising RGCs replacement therapy for restoring vision are impeded by insufficient RGC-like cells sources. The present work was enriched one new type RGC-like cells using two surface markers CD184 and CD171 from human induced pluripotent stem cells (hiPSCs) by FACS sorting firstly. These new kind cells have well proliferation ability and possessed passage tolerance in vitro 2D or 3D spheroids culture, which kept expressing Pax6, Brn3b and ßIII-Tubulin and so on. The transplanted CD184+CD171+ RGC-like cells could survive and integrate into the normal and optic nerve crush (ONC) mice retina, especially they were more inclined to across the optic nerve head and extend to the damaged optic nerve. These data support the feasible application for cell replacement therapy in RGC degenerative diseases, as well as help to develop new commercial cells sorting reagents and establish good manufacturing practice (GMP) grade RGC-like donor cells for further clinical application.


Assuntos
Células-Tronco Pluripotentes Induzidas , Traumatismos do Nervo Óptico , Camundongos , Animais , Humanos , Retina , Células Ganglionares da Retina , Nervo Óptico , Organoides , Modelos Animais de Doenças , Compressão Nervosa
3.
Int J Mol Sci ; 23(8)2022 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-35457277

RESUMO

Recent advances in the technology of producing novel cardiomyocytes from induced pluripotent stem cells (iPSC-cardiomyocytes) fuel new hope for future clinical applications. The use of iPSC-cardiomyocytes is particularly promising for the therapy of cardiac diseases such as myocardial infarction, where these cells could replace scar tissue and restore the functionality of the heart. Despite successful cardiogenic differentiation, medical applications of iPSC-cardiomyocytes are currently limited by their pronounced immature structural and functional phenotype. This review focuses on gap junction function in iPSC-cardiomyocytes and portrays our current understanding around the structural and the functional limitations of intercellular coupling and viable cardiac graft formation involving these novel cardiac muscle cells. We further highlight the role of the gap junction protein connexin 43 as a potential target for improving cell-cell communication and electrical signal propagation across cardiac tissue engineered from iPSC-cardiomyocytes. Better insight into the mechanisms that promote functional intercellular coupling is the foundation that will allow the development of novel strategies to combat the immaturity of iPSC-cardiomyocytes and pave the way toward cardiac tissue regeneration.


Assuntos
Transplante de Coração , Células-Tronco Pluripotentes Induzidas , Comunicação Celular/genética , Diferenciação Celular/fisiologia , Humanos , Miócitos Cardíacos/metabolismo , Doadores de Tecidos
4.
Am J Kidney Dis ; 78(3): 418-428, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33992729

RESUMO

Optimal glycemic control in kidney transplant recipients with diabetes is associated with improved morbidity and better patient and allograft survival. Transplant options for patients with diabetes requiring insulin therapy and chronic kidney disease who are suitable candidates for kidney transplantation should include consideration of ß-cell replacement therapy: pancreas or islet transplantation. International variation related to national regulatory policies exists in offering one or both options to suitable candidates and is further affected by pancreas/islet allocation policies and transplant waiting list dynamics. The selection of appropriate candidates depends on patient age, coexistent morbidities, the timing of referral to the transplant center (predialysis versus on dialysis) and availability of living kidney donors. Therefore, early referral (estimated glomerular filtration rate < 30 mL/min/1.73 m2) is of the utmost importance to ensure adequate time for informed decision making and thorough pretransplant evaluation. Obesity, cardiovascular disease, peripheral vascular disease, smoking, and frailty are some of the conditions that need to be addressed before acceptance on the transplant list, and ideally before dialysis becoming imminent. This review offers insights into selection of pancreas/islet transplant candidates by transplant centers and an update on posttransplant outcomes, which may have practice implications for referring nephrologists.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Nefropatias/cirurgia , Transplante de Rim/métodos , Doadores Vivos , Complicações Pós-Operatórias/epidemiologia , Saúde Global , Sobrevivência de Enxerto , Humanos , Morbidade/tendências , Transplante Homólogo
5.
Mol Biol (Mosk) ; 54(6): 939-954, 2020.
Artigo em Russo | MEDLINE | ID: mdl-33276357

RESUMO

Parkinson's disease is a widespread neurodegenerative disease, which is characterized by the death of dopaminergic neurons in the substantia nigra of the midbrain. Clinically, the disease is manifested by tremor, bradykinesia, muscle rigidity, and other motor and non-motor symptoms that ultimately lead to disability. To date, there are only symptomatic treatment options for Parkinson's disease; therefore, the search for new approaches is one of the most important directions of therapy for this disease. In the 1970's the idea of using cell replacement therapy based on the local nature and specificity of damage to a particular type of neuron in Parkinson's disease originated. The selection of the source of cells, the method and place of introduction, indications for this operation, and peculiarities of patient management have been in development for a long time. The efficiency of cell replacement therapy has been confirmed by a number of studies on animal models. Clinical trials have already begun and several more are planned soon. This review describes the main prerequisites for the use of cell replacement therapy in Parkinson's disease, the stages of development of this method, and clinical trials that have started in the last few years.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Doença de Parkinson , Animais , Neurônios Dopaminérgicos/patologia , Humanos , Mesencéfalo/patologia , Doença de Parkinson/terapia , Substância Negra/patologia
6.
Glia ; 67(2): 217-231, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30378163

RESUMO

Microglia are prominent immune cells in the central nervous system (CNS) and are critical players in both neurological development and homeostasis, and in neurological diseases when dysfunctional. Our previous understanding of the phenotypes and functions of microglia has been greatly extended by a dearth of recent investigations. Distinct genetically defined subsets of microglia are now recognized to perform their own independent functions in specific conditions. The molecular profiling of single microglial cells indicates extensively heterogeneous reactions in different neurological disorders, resulting in multiple potentials for crosstalk with other kinds of CNS cells such as astrocytes and neurons. In settings of neurological diseases it could thus be prudent to establish effective cell-based therapies by targeting entire microglial networks. Notably, activated microglial depletion through genetic targeting or pharmacological therapies within a suitable time window can stimulate replenishment of the CNS niche with new microglia. Additionally, enforced repopulation through provision of replacement cells also represents a potential means of exchanging dysfunctional with functional microglia. In each setting the newly repopulated microglia might have the potential to resolve ongoing neuroinflammation. In this review, we aim to summarize the most recent knowledge of microglia and to highlight microglial depletion and subsequent repopulation as a promising cell replacement therapy. Although glial cell replacement therapy is still in its infancy and future translational studies are still required, the approach is scientifically sound and provides new optimism for managing the neurotoxicity and neuroinflammation induced by activated microglia.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Microglia/fisiologia , Doenças do Sistema Nervoso/terapia , Animais , Microglia/patologia
7.
Eur J Neurosci ; 49(4): 472-486, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29923311

RESUMO

The dopamine precursor, levodopa, remains the "gold standard" treatment for Parkinson's disease, and, although it provides superlative efficacy in the early stages of the disease, its long-term use is limited by the development of severe motor side effects and a significant abating of therapeutic efficacy. Therefore, there remains a major unmet clinical need for the development of effective neuroprotective, neurorestorative or neuroreparatory therapies for this condition. The relatively selective loss of dopaminergic neurons from the nigrostriatal pathway makes Parkinson's disease an ideal candidate for reparative cell therapies, wherein the dopaminergic neurons that are lost in the condition are replaced through direct cell transplantation into the brain. To date, this approach has been developed, validated and clinically assessed using dopamine neuron-rich foetal ventral mesencephalon grafts which have been shown to survive and reinnervate the denervated brain after transplantation, and to restore motor function. However, despite long-term symptomatic relief in some patients, significant limitations, including poor graft survival and the impact this has on the number of foetal donors required, have prevented this therapy being more widely adopted as a restorative approach for Parkinson's disease. Injectable biomaterial scaffolds have the potential to improve the delivery, engraftment and survival of these grafts in the brain through provision of a supportive microenvironment for cell adhesion, growth and immune shielding. This article will briefly review the development of primary cell therapies for brain repair in Parkinson's disease and will consider the emerging literature which highlights the potential of using injectable biomaterial hydrogels in this context.


Assuntos
Materiais Biocompatíveis/uso terapêutico , Transplante de Tecido Fetal , Neurônios/transplante , Doença de Parkinson/terapia , Animais , Humanos
8.
Eur J Neurosci ; 49(4): 487-496, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30054941

RESUMO

Biomaterials have been shown to significantly improve the outcome of cellular reparative approaches for Parkinson's disease in experimental studies because of their ability to provide transplanted cells with a supportive microenvironment and shielding from the host immune system. However, given that the margin for improvement in such reparative therapies is considerable, further studies are required to fully investigate and harness the potential of biomaterials in this context. Given that several recent studies have demonstrated improved brain repair in Parkinsonian models when using dopaminergic grafts derived from younger foetal donors, we hypothesized that encapsulating these cells in a supportive biomaterial would further improve their reparative efficacy. Thus, this study aimed to determine the impact of a GDNF-loaded collagen hydrogel on the survival, reinnervation, and functional efficacy of dopaminergic neurons derived from young donors. To do so, hemi-Parkinsonian (6-hydroxydopamine-lesioned) rats received intrastriatal transplants of embryonic day 12 cells extracted from the rat ventral mesencephalon either alone, in a collagen hydrogel, with GDNF, or in a GDNF-loaded collagen hydrogel. Methamphetamine-induced rotational behaviour was assessed at three weekly intervals for a total of 12 weeks, after which rats were sacrificed for postmortem assessment of graft survival. We found that, following intrastriatal transplantation to the lesioned striatum, the GDNF-loaded collagen hydrogel significantly increased the survival (4-fold), reinnervation (5.4-fold), and functional efficacy of the embryonic day 12 dopaminergic neurons. In conclusion, this study further demonstrates the significant potential of biomaterial hydrogel scaffolds for cellular brain repair approaches in neurodegenerative diseases such as Parkinson's disease.


Assuntos
Materiais Biocompatíveis/uso terapêutico , Colágeno/uso terapêutico , Neurônios Dopaminérgicos/transplante , Transplante de Tecido Fetal/métodos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/uso terapêutico , Sobrevivência de Enxerto , Hidrogéis/uso terapêutico , Mesencéfalo/transplante , Neostriado/cirurgia , Doença de Parkinson/cirurgia , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Embrião de Mamíferos , Masculino , Oxidopamina , Ratos , Ratos Sprague-Dawley
9.
EMBO J ; 34(7): 841-55, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25733347

RESUMO

The discovery of insulin more than 90 years ago introduced a life-saving treatment for patients with type 1 diabetes, and since then, significant progress has been made in clinical care for all forms of diabetes. However, no method of insulin delivery matches the ability of the human pancreas to reliably and automatically maintain glucose levels within a tight range. Transplantation of human islets or of an intact pancreas can in principle cure diabetes, but this approach is generally reserved for cases with simultaneous transplantation of a kidney, where immunosuppression is already a requirement. Recent advances in cell reprogramming and beta cell differentiation now allow the generation of personalized stem cells, providing an unlimited source of beta cells for research and for developing autologous cell therapies. In this review, we will discuss the utility of stem cell-derived beta cells to investigate the mechanisms of beta cell failure in diabetes, and the challenges to develop beta cell replacement therapies. These challenges include appropriate quality controls of the cells being used, the ability to generate beta cell grafts of stable cellular composition, and in the case of type 1 diabetes, protecting implanted cells from autoimmune destruction without compromising other aspects of the immune system or the functionality of the graft. Such novel treatments will need to match or exceed the relative safety and efficacy of available care for diabetes.


Assuntos
Reprogramação Celular/imunologia , Terapia de Imunossupressão , Células Secretoras de Insulina , Transplante das Ilhotas Pancreáticas/imunologia , Imunologia de Transplantes , Animais , Autoenxertos , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/terapia , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/transplante , Transplante de Rim
10.
Mol Med ; 25(1): 9, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30922214

RESUMO

BACKGROUND: Cell therapy is proposed to be a potential treatment for Parkinson's disease (PD). Although fetal retinal pigment epithelial (RPE) cells have been tested in trials for treating PD patients, controversy has been raised over the issue of whether such cells can be reprogrammed into dopamine-producing cells for therapeutic efficacy. Here, we aim to investigate whether adult human RPE cells can be reprogrammed into dopamine-producing cells both in vitro and in the recipient monkey brain. METHODS: The RPE layer was isolated from frozen posterior eyeball tissue after penetrating keratoplasty surgery. The tumorigenicity of RPE cells was examined by G-banding and a tumor formation assay in nude mice. Immunogenicity was measured using a one-way mixed lymphocyte reaction (MLR) assay. Dopamine-production in chemically reprogrammed RPE cells was measured by HPLC. Finally, RPE cells were grafted into the brains of monkeys with MPTP-induced PD in order to investigate the potential of such cells treating PD patients in the future. RESULTS: RPE cell lines have been successively established from adult human eye tissues. Such cells can be chemically reprogrammed into dopamine-producing cells in vitro. Moreover, after being grafted into the brain caudate putamen of monkeys with MPTP-induced PD, RPE cells became tyrosine hydroxylase-positive cells, and recipient PD monkeys showed significant improvement of clinical conditions. CONCLUSIONS: This preclinical study using a primate model indicates that human adult RPE cells could be a potential cell source for the treatment of PD in the future.


Assuntos
Encéfalo/citologia , Terapia Baseada em Transplante de Células e Tecidos , Dopamina/metabolismo , Transtornos Parkinsonianos/terapia , Epitélio Pigmentado da Retina/citologia , Animais , Diferenciação Celular , Linhagem Celular , Humanos , Teste de Cultura Mista de Linfócitos , Macaca fascicularis , Masculino , Camundongos SCID , Pessoa de Meia-Idade , Epitélio Pigmentado da Retina/metabolismo
11.
Stem Cells ; 36(2): 146-160, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29178352

RESUMO

Two decades ago, researchers identified that a CAG expansion mutation in the huntingtin (HTT) gene was involved in the pathogenesis of Huntington's disease (HD). However, since the identification of the HTT gene, there has been no advance in the development of therapeutic strategies to prevent or reduce the progression of HD. With the recent advances in stem cell biology and human cell reprogramming technologies, several novel and exciting pathways have emerged allowing researchers to enhance their understanding of the pathogenesis of HD, to identify and screen potential drug targets, and to explore alternative donor cell sources for cell replacement therapy. This review will discuss the role of compensatory neurogenesis in the HD brain, the use of stem cell-based therapies for HD to replace or prevent cell loss, and the recent advance of cell reprogramming to model and/or treat HD. These new technologies, coupled with advances in genome editing herald a promising new era for HD research with the potential to identify a therapeutic strategy to alleviate this debilitating disorder. Stem Cells 2018;36:146-160.


Assuntos
Doença de Huntington/terapia , Animais , Edição de Genes , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Mutação/genética , Transplante de Células-Tronco
12.
Cell Immunol ; 326: 15-23, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28866278

RESUMO

Human pluripotent stem cells (hPSCs) promise a foreseeing future for regeneration medicine and cell replacement therapy with their abilities to produce almost any types of somatic cells of the body. The complicated immunogenicity of hPSC derivatives and context dependent responses in variable transplantations greatly hurdle the practical application of hPSCs in clinic. Especially for applications of hPSCs, induction of immune tolerance at the same time increases the risks of tumorigenesis. Over the past few years, thanks to the progress in immunology and practices in organ transplantation, endeavors on exploring strategies to induce long term protection of allogeneic transplants have shed light on overcoming this barrier. Novel genetic engineering techniques also allow to precisely cradle the immune response of transplantation. Here we reviewed the current understanding on immunogenicity, and efforts have been attempted on inducing immune tolerance for hPSC derivatives, with extra focus on modifying the graft cells. We also glimpse on employing cutting-edge genome editing technologies for this purpose, which will potentially endow hPSC derivatives with the nature of wide spectrum drugs for therapy.


Assuntos
Tolerância Imunológica/imunologia , Células-Tronco Pluripotentes/transplante , Transplante de Células-Tronco/métodos , Transplante de Células-Tronco/tendências , Imunologia de Transplantes/imunologia , Humanos , Células-Tronco Pluripotentes/imunologia
13.
J Neurosci ; 36(45): 11544-11558, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27911758

RESUMO

Chemogenetic manipulation of neuronal activities has been enabled by a designer receptor (designer receptor exclusively activated by designer drugs, DREADD) that is activated exclusively by clozapine-N-oxide (CNO). Here, we applied CNO as a functional reporter probe to positron emission tomography (PET) of DREADD in living brains. Mutant human M4 DREADD (hM4Di) expressed in transgenic (Tg) mouse neurons was visualized by PET with microdose [11C]CNO. Deactivation of DREADD-expressing neurons in these mice by nonradioactive CNO at a pharmacological dose could also be captured by arterial spin labeling MRI (ASL-MRI). Neural progenitors derived from hM4Di Tg-induced pluripotent stem cells were then implanted into WT mouse brains and neuronal differentiation of the grafts could be imaged by [11C]CNO-PET. Finally, ASL-MRI captured chemogenetic functional manipulation of the graft neurons. Our data provide the first demonstration of multimodal molecular/functional imaging of cells expressing a functional gene reporter in the brain, which would be translatable to humans for therapeutic gene transfers and cell replacements. SIGNIFICANCE STATEMENT: The present work provides the first successful demonstration of in vivo positron emission tomographic (PET) visualization of a chemogenetic designer receptor (designer receptor exclusively activated by designer drugs, DREADD) expressed in living brains. This technology has been applied to longitudinal PET reporter imaging of neuronal grafts differentiated from induced pluripotent stem cells. Differentiated from currently used reporter genes for neuroimaging, DREADD has also been available for functional manipulation of target cells, which could be visualized by functional magnetic resonance imaging (fMRI) in a real-time manner. Multimodal imaging with PET/fMRI enables the visualization of the differentiation of iPSC-derived neural progenitors into mature neurons and DREADD-mediated functional manipulation along the time course of the graft and is accordingly capable of fortifying the utility of stem cells in cell replacement therapies.


Assuntos
Encéfalo/citologia , Genes Reporter , Células-Tronco Pluripotentes Induzidas/citologia , Imagem Multimodal/métodos , Células-Tronco Neurais/transplante , Neurônios/citologia , Neurônios/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Tomografia por Emissão de Pósitrons/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Transplante de Células-Tronco/métodos
14.
Dev Biol ; 417(2): 229-51, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27059883

RESUMO

Over the last 20 years, there has been increasing focus on the development of novel stem cell based therapies for the treatment of disorders and diseases affecting the enteric nervous system (ENS) of the gastrointestinal tract (so-called enteric neuropathies). Here, the idea is that ENS progenitor/stem cells could be transplanted into the gut wall to replace the damaged or absent neurons and glia of the ENS. This White Paper sets out experts' views on the commonly used methods and approaches to identify, isolate, purify, expand and optimize ENS stem cells, transplant them into the bowel, and assess transplant success, including restoration of gut function. We also highlight obstacles that must be overcome in order to progress from successful preclinical studies in animal models to ENS stem cell therapies in the clinic.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Sistema Nervoso Entérico/patologia , Trato Gastrointestinal/patologia , Doença de Hirschsprung/terapia , Pseudo-Obstrução Intestinal/terapia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Animais , Modelos Animais de Doenças , Trato Gastrointestinal/inervação , Guias como Assunto , Doença de Hirschsprung/patologia , Humanos , Pseudo-Obstrução Intestinal/patologia
15.
Diabetologia ; 59(9): 1838-42, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27412250

RESUMO

Pancreatic beta cells differ in terms of glucose responsiveness, insulin secretion and proliferative capacity; however, the molecular pathways that regulate this cellular heterogeneity are unknown. We have identified the Wnt-planar cell polarity (PCP) effector Flattop (FLTP) as a biomarker that identifies mature beta cells in the islets of Langerhans. Interestingly, three-dimensional architecture and Wnt-PCP ligands are sufficient to trigger mouse and human beta cell maturation. These results highlight the fact that novel biomarkers shed light on the long-standing mystery of beta cell heterogeneity and identify the Wnt-PCP pathway as triggering beta cell maturation. Understanding heterogeneity in the islets of Langerhans might allow targeting of beta cell subpopulations for regenerative therapy and provide building principles for stem cell-derived islets. This review summarises a presentation given at the 'Can we make a better beta cell?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Amin Ardestani and Kathrin Maedler, DOI: 10.1007/s00125-016-3892-9 , and by Harry Heimberg and colleagues, DOI: 10.1007/s00125-016-3879-6 ) and a commentary by the Session Chair, Shanta Persaud (DOI: 10.1007/s00125-016-3870-2 ).


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos , Ilhotas Pancreáticas/citologia , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Via de Sinalização Wnt/fisiologia
16.
Neurochem Res ; 41(6): 1238-49, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26758268

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder that is caused by a loss of dopaminergic (DAergic) neurons in mesencephalic substantia nigra (SN). Human umbilical cord mesenchymal stem cells (hUC-MSCs) are capable of self-renewal and differentiation into multiple cell lineages, including DAergic neurons. Thus, hUC-MSCs could be a promising alternative to compensate for the loss of DAergic neurons in PD. In the current study, hUC-MSCs and hUC-MSCs-derived DAergic-like neurons were transplanted into the striatum and SN of a rat model of PD that is induced by 6-hydroxydopamine (6-OHDA). We evaluated their therapeutic effects on improving rotation behavior in the rat and on modulating the level of heat shock protein 60 (Hsp60) expression in the brain. After transplantation, an amelioration of rotation behavior was observed in rats that underwent cell grafting, and hUC-MSCs-derived DAergic-like neurons were superior to hUC-MSCs at inducing behavioral improvement. Western blot and immunohistochemistry analysis indicated significantly elevated levels of Hsp60 in cell-grafted rats compared to 6-OHDA-lesioned (PD) rats. These results demonstrate that hUC-MSCs-based cell transplantation is potential therapeutic treatment for PD, and hUC-MSCs-derived DAergic-like neurons appear to be favorable candidates for cell replacement therapy in PD. Finally, Hsp60 could be involved in a mechanism of behavioral recovery.


Assuntos
Chaperonina 60/biossíntese , Neurônios Dopaminérgicos/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Proteínas Mitocondriais/biossíntese , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/terapia , Cordão Umbilical/transplante , Animais , Comportamento Animal/fisiologia , Células Cultivadas , Corpo Estriado/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Cordão Umbilical/citologia
17.
J Neurochem ; 130(4): 472-89, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24773031

RESUMO

Dopamine replacement therapy in Parkinson's disease is associated with several unwanted effects, of which dyskinesia is the most disabling. The development of new therapeutic interventions to reduce the impact of dyskinesia in Parkinson's disease is therefore a priority need. This review summarizes the key molecular mechanisms that underlie dyskinesia. The role of dopamine receptors and their associated signaling mechanisms including dopamine-cAMP-regulated neuronal phosphoprotein, extracellular signal-regulated kinase, mammalian target of rapamycin, mitogen and stress-activated kinase-1 and Histone H3 are summarized, along with an evaluation of the role of cannabinoid and nicotinic acetylcholine receptors. The role of synaptic plasticity and animal behavioral results on dyskinesia are also evaluated. The most recent therapeutic advances to treat Parkinson's disease are discussed, with emphasis on the possibilities and limitations of non-pharmacological interventions such as physical activity, deep brain stimulation, transcranial magnetic field stimulation and cell replacement therapy. The review suggests new prospects for the management of Parkinson's disease-associated motor symptoms, especially the development of dyskinesia. This review aims at summarizing the key molecular mechanisms underlying dyskinesia and the most recent therapeutic advances to treat Parkinson's disease with emphasis on non-pharmacological interventions such as physical activity, deep brain stimulation (DBS), transcranial magnetic field stimulation (TMS) and cell replacement therapy. These new interventions are discussed from both the experimental and clinical point of view, describing their current strength and limitations.


Assuntos
Discinesias/etiologia , Discinesias/terapia , Doença de Parkinson/complicações , Doença de Parkinson/terapia , Animais , Canabinoides/farmacologia , Transplante de Células , Cromatina/efeitos dos fármacos , Estimulação Encefálica Profunda , Dopaminérgicos/uso terapêutico , Fosfoproteína 32 Regulada por cAMP e Dopamina/fisiologia , Discinesias/fisiopatologia , Histonas/metabolismo , Humanos , Levodopa/farmacologia , Levodopa/uso terapêutico , Atividade Motora/fisiologia , Doença de Parkinson/fisiopatologia , Fosforilação , Desempenho Psicomotor/fisiologia , Receptores de Dopamina D1/efeitos dos fármacos , Receptores de Dopamina D1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estimulação Magnética Transcraniana
18.
J Neural Eng ; 21(5)2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39231475

RESUMO

Objective.Cryogel microcarriers made of poly(ethylene glycol) diacrylate and 3-sulfopropyl acrylate have the potential to act as delivery vehicles for long-term retention of neurotrophic factors (NTFs) in the brain. In addition, they can potentially enhance stem cell-derived dopaminergic (DAergic) cell replacement strategies for Parkinson's disease (PD), by addressing the limitations of variable survival and poor differentiation of the transplanted precursors due to neurotrophic deprivation post-transplantation in the brain. In this context, to develop a proof-of-concept, the aim of this study was to determine the efficacy of glial cell line-derived NTF (GDNF)-loaded cryogel microcarriers by assessing their impact on the survival of, and reinnervation by, primary DAergic grafts after intra-striatal delivery in Parkinsonian rat brains.Approach.Rat embryonic day 14 ventral midbrain cells were transplanted into the 6-hydroxydopamine-lesioned striatum either alone, or with GDNF, or with unloaded cryogel microcarriers, or with GDNF-loaded cryogel microcarriers.Post-mortem, GDNF and tyrosine hydroxylase immunostaining were used to identify retention of the delivered GDNF within the implanted cryogel microcarriers, and to identify the transplanted DAergic neuronal cell bodies and fibres in the brains, respectively.Main results.We found an intact presence of GDNF-stained cryogel microcarriers in graft sites, indicating their ability for long-term retention of the delivered GDNF up to 4 weeks in the brain. This resulted in an enhanced survival (1.9-fold) of, and striatal reinnervation (density & volume) by, the grafted DAergic neurons, in addition to an enhanced sprouting of fibres within graft sites.Significance.This data provides an important proof-of-principle for the beneficial effects of neurotrophin-loaded cryogel microcarriers on engraftment of cells in the context of cell replacement therapy in PD. For clinical translation, further studies will be needed to assess the impact of cryogel microcarriers on the survival and differentiation of stem cell-derived DAergic precursors in Parkinsonian rat brains.


Assuntos
Criogéis , Neurônios Dopaminérgicos , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Animais , Fator Neurotrófico Derivado de Linhagem de Célula Glial/administração & dosagem , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Ratos , Criogéis/administração & dosagem , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/transplante , Doença de Parkinson/terapia , Ratos Sprague-Dawley , Modelos Animais de Doenças , Células Cultivadas , Masculino
19.
J Parkinsons Dis ; 14(3): 415-435, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38457149

RESUMO

Parkinson's disease (PD) is an increasingly prevalent neurological disorder, affecting more than 8.5 million individuals worldwide. α-Synucleinopathy in PD is considered to cause dopaminergic neuronal loss in the substantia nigra, resulting in characteristic motor dysfunction that is the target for current medical and surgical therapies. Standard treatment for PD has remained unchanged for several decades and does not alter disease progression. Furthermore, symptomatic therapies for PD are limited by issues surrounding long-term efficacy and side effects. Cell replacement therapy (CRT) presents an alternative approach that has the potential to restore striatal dopaminergic input and ameliorate debilitating motor symptoms in PD. Despite promising pre-clinical data, CRT has demonstrated mixed success clinically. Recent advances in graft biology have renewed interest in the field, resulting in several worldwide ongoing clinical trials. However, factors surrounding the effective neurosurgical delivery of cell grafts have remained under-studied, despite their significant potential to influence therapeutic outcomes. Here, we focus on the key neurosurgical factors to consider for the clinical translation of CRT. We review the instruments that have been used for cell graft delivery, highlighting current features and limitations, while discussing how future devices could address these challenges. Finally, we review other novel developments that may enhance graft accessibility, delivery, and efficacy. Challenges surrounding neurosurgical delivery may critically contribute to the success of CRT, so it is crucial that we address these issues to ensure that CRT does not falter at the final hurdle.


Assuntos
Neurônios Dopaminérgicos , Doença de Parkinson , Humanos , Doença de Parkinson/terapia , Animais
20.
Artigo em Inglês | MEDLINE | ID: mdl-38747224

RESUMO

Alzheimer's disease (AD), an inexorable neurodegenerative ailment marked by cognitive impairment and neuropsychiatric manifestations, stands as the foremost prevailing form of dementia in the geriatric population. Its pathological signs include the aggregation of amyloid proteins, hyperphosphorylation of tau proteins, and the consequential loss of neural cells. The etiology of AD has prompted the formulation of numerous conjectures, each endeavoring to elucidate its pathogenesis. While a subset of therapeutic agents has displayed clinical efficacy in AD patients, a significant proportion has encountered disappointment. Notably, the extent of neural cell depletion bears a direct correlation with the disease's progressive severity. However, the absence of efficacious therapeutic remedies for neurodegenerative afflictions engenders a substantial societal burden and exerts a notable economic toll. In the past two decades, the realm of regenerative cell therapy, referred to as stem cell therapy, has unfolded as an avenue for the exploration of profoundly innovative approaches to treat neurodegenerative conditions. This promise is underpinned by the remarkable capacity of stem cells to remediate compromised neural tissue by means of cell replacement, to cultivate an environment conducive to regeneration, and to shield extant healthy neuronal and glial components from further degradation. Thus, this review aims to delve into the current knowledge of stem cell-based therapies and future possibilities in this domain.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA