Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Stem Cells Dev ; 33(9-10): 201-213, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38390839

RESUMO

Because derivation of retinal organoids (ROs) and transplantation are frequently split between geographically distant locations, we developed a special shipping device and protocol capable of the organoids' delivery to any location. Human embryonic stem cell (hESC)-derived ROs were differentiated from the hESC line H1 (WA01), shipped overnight to another location, and then transplanted into the subretinal space of blind immunodeficient retinal degeneration (RD) rats. Development of transplants was monitored by spectral-domain optical coherence tomography. Visual function was accessed by optokinetic tests and superior colliculus (SC) electrophysiology. Cryostat sections through transplants were stained with hematoxylin and eosin; or processed for immunohistochemistry to label human donor cells, retinal cell types, and synaptic markers. After transplantation, ROs integrated into the host RD retina, formed functional photoreceptors, and improved vision in rats with advanced RD. The survival and vision improvement are comparable with our previous results of hESC-ROs without a long-distance delivery. Furthermore, for the first time in the stem cell transplantation field, we demonstrated that the response heatmap on the SC showed a similar shape to the location of the transplant in the host retina, which suggested the point-to-point projection of the transplant from the retina to SC. In conclusion, our results showed that using our special device and protocol, the hESC-derived ROs can be shipped over long distance and are capable of survival and visual improvement after transplantation into the RD rats. Our data provide a proof-of-concept for stem cell replacement as a therapy for RD patients.


Assuntos
Células-Tronco Embrionárias Humanas , Organoides , Retina , Degeneração Retiniana , Animais , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/transplante , Degeneração Retiniana/terapia , Degeneração Retiniana/patologia , Humanos , Organoides/citologia , Organoides/transplante , Ratos , Retina/citologia , Retina/patologia , Diferenciação Celular , Transplante de Células-Tronco/métodos , Sobrevivência Celular , Tomografia de Coerência Óptica
2.
Ophthalmology ; 131(6): 682-691, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38160882

RESUMO

PURPOSE: To report long-term results from a phase 1/2a clinical trial assessment of a scaffold-based human embryonic stem cell-derived retinal pigmented epithelium (RPE) implant in patients with advanced geographic atrophy (GA). DESIGN: A single-arm, open-label phase 1/2a clinical trial approved by the United States Food and Drug Administration. PARTICIPANTS: Patients were 69-85 years of age at the time of enrollment and were legally blind in the treated eye (best-corrected visual acuity [BCVA], ≤ 20/200) as a result of GA involving the fovea. METHODS: The clinical trial enrolled 16 patients, 15 of whom underwent implantation successfully. The implant was administered to the worse-seeing eye with the use of a custom subretinal insertion device. The companion nonimplanted eye served as the control. The primary endpoint was at 1 year; thereafter, patients were followed up at least yearly. MAIN OUTCOME MEASURES: Safety was the primary endpoint of the study. The occurrence and frequency of adverse events (AEs) were determined by scheduled eye examinations, including measurement of BCVA and intraocular pressure and multimodal imaging. Serum antibody titers were collected to monitor systemic humoral immune responses to the implanted cells. RESULTS: At a median follow-up of 3 years, fundus photography revealed no migration of the implant. No unanticipated, severe, implant-related AEs occurred, and the most common anticipated severe AE (severe retinal hemorrhage) was eliminated in the second cohort (9 patients) through improved intraoperative hemostasis. Nonsevere, transient retinal hemorrhages were noted either during or after surgery in all patients as anticipated for a subretinal surgical procedure. Throughout the median 3-year follow-up, results show that implanted eyes were more likely to improve by > 5 letters of BCVA and were less likely to worsen by > 5 letters compared with nonimplanted eyes. CONCLUSIONS: This report details the long-term follow-up of patients with GA to receive a scaffold-based stem cell-derived bioengineered RPE implant. Results show that the implant, at a median 3-year follow-up, is safe and well tolerated in patients with advanced dry age-related macular degeneration. The safety profile, along with the early indication of efficacy, warrants further clinical evaluation of this novel approach for the treatment of GA. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.


Assuntos
Atrofia Geográfica , Epitélio Pigmentado da Retina , Acuidade Visual , Humanos , Atrofia Geográfica/cirurgia , Atrofia Geográfica/fisiopatologia , Epitélio Pigmentado da Retina/transplante , Epitélio Pigmentado da Retina/patologia , Idoso , Acuidade Visual/fisiologia , Feminino , Idoso de 80 Anos ou mais , Masculino , Seguimentos , Tomografia de Coerência Óptica , Células-Tronco Embrionárias Humanas/transplante , Células-Tronco Embrionárias Humanas/citologia , Transplante de Células-Tronco , Resultado do Tratamento
3.
Tissue Eng Regen Med ; 19(6): 1349-1358, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36036887

RESUMO

BACKGROUND: Spinal cord injury (SCI) results in permanent impairment of motor and sensory functions at and below the lesion site. There is no therapeutic option to the functional recovery of SCI involving diverse injury responses of different cell types in the lesion that limit endogenous nerve regeneration. In this regard, cell replacement therapy utilizing stem cells or their derivatives has become a highly promising approach to promote locomotor recovery. For this reason, the demand for a safe and efficient multipotent cell source that can differentiate into various neural cells is increasing. In this study, we evaluated the efficacy and safety of human polysialylated-neural cell adhesion molecule (PSA-NCAM)-positive neural precursor cells (hNPCsPSA-NCAM+) as a treatment for SCI. METHODS: One hundred thousand hNPCsPSA-NCAM+ isolated from human embryonic stem cell-derived NPCs were transplanted into the lesion site by microinjection 7 days after contusive SCI at the thoracic level. We examined the histological characteristics of the graft and behavioral improvement in the SCI rats 10 weeks after transplantation. RESULTS: Locomotor activity improvement was estimated by the Basso-Beattie-Bresnahan locomotor rating scale. Behavioral tests revealed that the transplantation of the hNPCsPSA-NCAM+ into the injured spinal cords of rats significantly improved locomotor function. Histological examination showed that hNPCsPSA-NCAM+ had differentiated into neural cells and successfully integrated into the host tissue with no evidence of tumor formation. We investigated cytokine expressions, which led to the early therapeutic effect of hNPCsPSA-NCAM+, and found that some undifferentiated NPCs still expressed midkine, a well-known neurotrophic factor involved in neural development and inflammatory responses, 10 weeks after transplantation. CONCLUSION: Our results demonstrate that hNPCsPSA-NCAM+ serve as a safe and efficient cell source which has the potential to improve impaired motor function following SCI.


Assuntos
Células-Tronco Embrionárias Humanas , Células-Tronco Neurais , Traumatismos da Medula Espinal , Ratos , Animais , Humanos , Moléculas de Adesão de Célula Nervosa/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/patologia , Células-Tronco Embrionárias Humanas/transplante , Traumatismos da Medula Espinal/terapia , Modelos Animais de Doenças
4.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35163361

RESUMO

Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach for diverse diseases and injuries. The biological and clinical advantages of human fetal MSCs (hfMSCs) have recently been reported. In terms of promising therapeutic approaches for diverse diseases and injuries, hfMSCs have gained prominence as healing tools for clinical therapies. Therefore, this review assesses not the only biological advantages of hfMSCs for healing human diseases and regeneration, but also the research evidence for the engraftment and immunomodulation of hfMSCs based on their sources and biological components. Of particular clinical relevance, the present review also suggests the potential therapeutic feasibilities of hfMSCs for musculoskeletal disorders, including osteoporosis, osteoarthritis, and osteogenesis imperfecta.


Assuntos
Células-Tronco Embrionárias Humanas/transplante , Doenças Musculoesqueléticas/terapia , Animais , Humanos , Imunomodulação , Doenças Musculoesqueléticas/imunologia , Resultado do Tratamento
5.
Sci Rep ; 11(1): 18921, 2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34584110

RESUMO

Articular cartilage functions as a shock absorber and facilitates the free movement of joints. Currently, there are no therapeutic drugs that promote the healing of damaged articular cartilage. Limitations associated with the two clinically relevant cell populations, human articular chondrocytes and mesenchymal stem cells, necessitate finding an alternative cell source for cartilage repair. Human embryonic stem cells (hESCs) provide a readily accessible population of self-renewing, pluripotent cells with perceived immunoprivileged properties for cartilage generation. We have developed a robust method to generate 3D, scaffold-free, hyaline cartilage tissue constructs from hESCs that are composed of numerous chondrocytes in lacunae, embedded in an extracellular matrix containing Type II collagen, sulphated glycosaminoglycans and Aggrecan. The elastic (Young's) modulus of the hESC-derived cartilage tissue constructs (0.91 ± 0.08 MPa) was comparable to full-thickness human articular cartilage (0.87 ± 0.09 MPa). Moreover, we have successfully scaled up the size of the scaffold-free, 3D hESC-derived cartilage tissue constructs to between 4.5 mm and 6 mm, thus enhancing their suitability for clinical application.


Assuntos
Cartilagem Articular/crescimento & desenvolvimento , Células-Tronco Embrionárias Humanas/metabolismo , Engenharia Tecidual/métodos , Agrecanas/metabolismo , Cartilagem/metabolismo , Cartilagem Articular/metabolismo , Diferenciação Celular , Células Cultivadas , Condrócitos/metabolismo , Condrogênese , Colágeno Tipo II/metabolismo , Matriz Extracelular/metabolismo , Glicosaminoglicanos/metabolismo , Regeneração Tecidual Guiada/métodos , Células-Tronco Embrionárias Humanas/transplante , Humanos , Células-Tronco Mesenquimais/metabolismo
6.
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
7.
Stem Cells Transl Med ; 10(11): 1491-1499, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34302450

RESUMO

Experimental cell models are indispensable for clarifying the pathophysiology of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and for developing therapeutic agents. To recapitulate the symptoms and drug response of COVID-19 patients in vitro, SARS-CoV-2 studies using physiologically relevant human embryonic stem (ES)/induced pluripotent stem (iPS) cell-derived somatic cells and organoids are ongoing. These cells and organoids have been used to show that SARS-CoV-2 can infect and damage various organs including the lung, heart, brain, intestinal tract, kidney, and pancreas. They are also being used to develop COVID-19 therapeutic agents, including evaluation of their antiviral efficacy and safety. The relationship between COVID-19 aggravation and human genetic backgrounds has been investigated using genetically modified ES/iPS cells and patient-derived iPS cells. This review summarizes the latest results and issues of SARS-CoV-2 research using human ES/iPS cell-derived somatic cells and organoids.


Assuntos
COVID-19 , Células-Tronco Embrionárias Humanas/fisiologia , Organoides/fisiologia , SARS-CoV-2/fisiologia , Pesquisa Biomédica/métodos , Pesquisa Biomédica/tendências , COVID-19/etiologia , COVID-19/patologia , COVID-19/terapia , Terapia Genética/métodos , Terapia Genética/tendências , Células-Tronco Embrionárias Humanas/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Organoides/citologia , Organoides/transplante
8.
Cells ; 10(5)2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-34066487

RESUMO

The worldwide rise in prevalence of chronic kidney disease (CKD) demands innovative bio-medical solutions for millions of kidney patients. Kidney regenerative medicine aims to replenish tissue which is lost due to a common pathological pathway of fibrosis/inflammation and rejuvenate remaining tissue to maintain sufficient kidney function. To this end, cellular therapy strategies devised so far utilize kidney tissue-forming cells (KTFCs) from various cell sources, fetal, adult, and pluripotent stem-cells (PSCs). However, to increase engraftment and potency of the transplanted cells in a harsh hypoxic diseased environment, it is of importance to co-transplant KTFCs with vessel forming cells (VFCs). VFCs, consisting of endothelial cells (ECs) and mesenchymal stem-cells (MSCs), synergize to generate stable blood vessels, facilitating the vascularization of self-organizing KTFCs into renovascular units. In this paper, we review the different sources of KTFCs and VFCs which can be mixed, and report recent advances made in the field of kidney regeneration with emphasis on generation of vascularized kidney tissue by cell transplantation.


Assuntos
Células Endoteliais/transplante , Células-Tronco Embrionárias Humanas/transplante , Transplante de Células-Tronco Mesenquimais , Medicina Regenerativa/métodos , Insuficiência Renal Crônica/terapia , Manejo de Espécimes/métodos , Animais , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/transplante
9.
Stem Cell Reports ; 16(4): 851-867, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33770500

RESUMO

ESC- and iPSC-derived retinal transplantation is a promising therapeutic approach for disease with end-stage retinal degeneration, such as retinitis pigmentosa and age-related macular degeneration. We previously showed medium- to long-term survival, maturation, and light response of transplanted human ESC- and iPSC-retina in mouse, rat, and monkey models of end-stage retinal degeneration. Because the use of patient hiPSC-derived retina with a disease-causing gene mutation is not appropriate for therapeutic use, allogeneic transplantation using retinal tissue/cells differentiated from a stocked hESC and iPSC line would be most practical. Here, we characterize the immunological properties of hESC- and iPSC-retina and present their three major advantages: (1) hESC- and iPSC-retina expressed low levels of human leukocyte antigen (HLA) class I and little HLA class II in vitro, (2) hESC- and iPSC-retina greatly suppressed immune activation of lymphocytes in co-culture, and (3) hESC- and iPSC-retina suppressed activated immune cells partially via transforming growth factor ß signaling. These results support the use of allogeneic hESC- and iPSC-retina in future clinical application.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Humanas/citologia , Terapia de Imunossupressão , Células-Tronco Pluripotentes Induzidas/citologia , Retina/imunologia , Animais , Diferenciação Celular/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/transplante , Humanos , Imunomodulação/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Interferon gama/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Primatas , Proteínas Recombinantes/farmacologia , Epitélio Pigmentado da Retina/citologia , Fator de Crescimento Transformador beta/metabolismo
10.
Artif Organs ; 45(5): 447-453, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33590913

RESUMO

Recent advances in developmental biology and stem cell biology have led to the increased availability of extrarenal stem cells, including mesenchymal/stromal stem cells (MSCs), renal stem or progenitor cells isolated from embryonic and adult kidneys, and kidney lineage cells or tissues generated from human pluripotent stem cells (hPSCs), such as human embryonic stem cells and human-induced pluripotent stem cells. Regenerative medicine strategies for kidney diseases are largely categorized into the transplantation of reconstructed kidney organs and cell therapies. Reconstruction is being attempted by hPSC-derived kidney lineage cells with various strategies, such as self-organization, interspecies blastocyst complementation, utilization of a xenogeneic organ niche, decellularization and repopulation, and 3D bioprinting. However, cell therapies using extrarenal stem cells, such as MSCs, and renal stem or progenitor cells derived from embryonic and adult kidneys or differentiated from hPSCs have been investigated in animal models of both acute kidney injury and chronic kidney disease. Indeed, multiple clinical trials using MSCs, bone marrow stem cells, and kidney-derived cells have already been carried out. This review summarizes the current status and future perspective of kidney regenerative medicine strategies and discusses the closest and fastest strategies to solving the medical and economic problems associated with kidney diseases.


Assuntos
Transplante de Rim/métodos , Medicina Regenerativa/métodos , Insuficiência Renal Crônica/terapia , Animais , Bioimpressão/métodos , Bioimpressão/tendências , Diferenciação Celular , Efeitos Psicossociais da Doença , Modelos Animais de Doenças , Células-Tronco Embrionárias Humanas/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Rim/citologia , Rim/fisiopatologia , Transplante de Células-Tronco Mesenquimais , Medicina Regenerativa/tendências , Insuficiência Renal Crônica/economia , Insuficiência Renal Crônica/fisiopatologia
11.
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
12.
Cell Rep ; 33(10): 108455, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33296648

RESUMO

The ever-increasing therapeutic and pharmaceutical demand for liver cells calls for systems that enable mass production of hepatic cells. Here we describe a large-scale suspension system that uses human endoderm stem cells (hEnSCs) as precursors to generate functional and transplantable hepatocytes (E-heps) or cholangiocytes (E-chos). hEnSC-derived hepatic populations are characterized by single-cell transcriptomic analyses and compared with hESC-derived counterparts, in-vitro-maintained or -expanded primary hepatocytes and adult cells, which reveals that hepatic differentiation of hEnSCs recapitulates in vivo development and that the heterogeneities of the resultant populations can be manipulated by regulating the EGF and MAPK signaling pathways. Functional assessments demonstrate that E-heps and E-chos possess properties comparable with adult counterparts and that, when transplanted intraperitoneally, encapsulated E-heps were able to rescue rats with acute liver failure. Our study lays the foundation for cell-based therapeutic agents and in vitro applications for liver diseases.


Assuntos
Técnicas de Cultura de Células/métodos , Endoderma/citologia , Hepatócitos/citologia , Células-Tronco Embrionárias Humanas/citologia , Ductos Biliares/citologia , Ductos Biliares/metabolismo , Diferenciação Celular/fisiologia , Endoderma/metabolismo , Endoderma/transplante , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Hepatócitos/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/transplante , Humanos , Fígado/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante
13.
J Mater Chem B ; 8(40): 9277-9294, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-32996553

RESUMO

Impaired skin regeneration in chronic wounds like in diabetes corresponds to high oxidative stress, poor angiogenesis and insufficient collagen hyperplasia. Therefore, a multifaceted strategy for treatment is required to address critical issues associated with chronic wound healing. Fascinating application of nanomaterials in chronic wounds is still limited; hence, in the present work bioactive solubilized decellularized dermal matrix (sADM) was employed to form a hydrogel with chitosan (CTS) at physiological pH/temperature and modified with reactive oxygen species (ROS) scavenging carbon nanodots (ND). A detailed in vitro investigation found that the ND modified bioactive hydrogel (CsADMND) is suitable for human amniotic membrane derived stem cell (hAMSC) delivery. Also, CsADMND was observed to possess a good ROS scavenging property, hemocompatibility and pro-angiogenic potential as demonstrated by 2,2-diphenyl-1-picrylhydrazyl (DPPH), haemolysis and chick chorioallantoic membrane (CAM) assay, respectively. The hybrid hydrogel promoted migration of cells in vitro in scratch assay owing to its antioxidant potential and the presence of bioactive moieties. Further, its efficacy in healing full thickness (FT) chronic wounds was evaluated in a streptozotocin (STZ) induced diabetic model. The CsADMND hydrogel after association with hAMSCs led to stimulation of early angiogenesis, superior collagen deposition, rapid wound closure, complete reepithelialisation, and formation of distinct organized dermal epidermal junctions (DEJ) post 21 days of healing. These results suggest that the hAMSC laden CsADMND hydrogel may serve as a promising therapeutic strategy for the management of chronic wounds.


Assuntos
Derme Acelular , Células-Tronco Embrionárias Humanas/transplante , Hidrogéis/química , Pontos Quânticos/uso terapêutico , Cicatrização/efeitos dos fármacos , Âmnio/citologia , Animais , Antibacterianos/química , Antibacterianos/uso terapêutico , Carbono/química , Quitosana/química , Diabetes Mellitus Experimental/fisiopatologia , Escherichia coli/efeitos dos fármacos , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/uso terapêutico , Humanos , Masculino , Testes de Sensibilidade Microbiana , Neovascularização Fisiológica/efeitos dos fármacos , Pontos Quânticos/química , Ratos Wistar , Reepitelização/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos
14.
PLoS One ; 15(8): e0237426, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32813731

RESUMO

Transplantation of human embryonic stem cell (hESC)-derived neural progenitors is a potential treatment for neurological disorders, but relatively little is known about the time course for human neuron maturation after transplantation and the emergence of morphological and electrophysiological properties. To address this gap, we transplanted hESC-derived human GABAergic interneuron progenitors into the mouse hippocampus, and then characterized their electrophysiological properties and dendritic arborizations after transplantation by means of ex vivo whole-cell patch clamp recording, followed by biocytin staining, confocal imaging and neuron reconstruction software. We asked whether particular electrophysiological and morphological properties showed maturation-dependent changes after transplantation. We also investigated whether the emergence of particular electrophysiological properties were linked to increased complexity of the dendritic arbors. Human neurons were classified into five distinct neuronal types (Type I-V), ranging from immature to mature fast-spiking interneurons. Hierarchical clustering of the dendritic morphology and Sholl analyses suggested four morphologically distinct classes (Class A-D), ranging from simple/immature to highly complex. Incorporating all of our data regardless of neuronal classification, we investigated whether any electrophysiological and morphological features correlated with time post-transplantation. This analysis demonstrated that both dendritic arbors and electrophysiological properties matured after transplantation.


Assuntos
Fenômenos Eletrofisiológicos , Hipocampo/citologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/transplante , Interneurônios/citologia , Interneurônios/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Estudos de Viabilidade , Feminino , Humanos , Masculino , Camundongos
15.
Cell Death Dis ; 11(5): 354, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32393784

RESUMO

Human pluripotent stem cells (hPSCs)-derived cardiovascular progenitor cells (CVPCs) are a promising source for myocardial repair, while the mechanisms remain largely unknown. Extracellular vesicles (EVs) are known to mediate cell-cell communication, however, the efficacy and mechanisms of hPSC-CVPC-secreted EVs (hCVPC-EVs) in the infarct healing when given at the acute phase of myocardial infarction (MI) are unknown. Here, we report the cardioprotective effects of the EVs secreted from hESC-CVPCs under normoxic (EV-N) and hypoxic (EV-H) conditions in the infarcted heart and the long noncoding RNA (lncRNA)-related mechanisms. The hCVPC-EVs were confirmed by electron microscopy, nanoparticle tracking, and immunoblotting analysis. Injection of hCVPC-EVs into acutely infracted murine myocardium significantly improved cardiac function and reduced fibrosis at day 28 post MI, accompanied with the improved vascularization and cardiomyocyte survival at border zones. Consistently, hCVPC-EVs enhanced the tube formation and migration of human umbilical vein endothelial cells (HUVECs), improved the cell viability, and attenuated the lactate dehydrogenase release of neonatal rat cardiomyocytes (NRCMs) with oxygen glucose deprivation (OGD) injury. Moreover, the improvement of the EV-H in cardiomyocyte survival and tube formation of HUVECs was significantly better than these in the EV-N. RNA-seq analysis revealed a high abundance of the lncRNA MALAT1 in the EV-H. Its abundance was upregulated in the infarcted myocardium and cardiomyocytes treated with hCVPC-EVs. Overexpression of human MALAT1 improved the cell viability of NRCM with OGD injury, while knockdown of MALAT1 inhibited the hCVPC-EV-promoted tube formation of HUVECs. Furthermore, luciferase activity assay, RNA pull-down, and manipulation of miR-497 levels showed that MALAT1 improved NRCMs survival and HUVEC tube formation through targeting miR-497. These results reveal that hCVPC-EVs promote the infarct healing through improvement of cardiomyocyte survival and angiogenesis. The cardioprotective effects of hCVPC-EVs can be enhanced by hypoxia-conditioning of hCVPCs and are partially contributed by MALAT1 via targeting the miRNA.


Assuntos
Vesículas Extracelulares/transplante , Células-Tronco Embrionárias Humanas/transplante , Infarto do Miocárdio/cirurgia , Miocárdio/metabolismo , Miócitos Cardíacos/transplante , Função Ventricular Esquerda , Remodelação Ventricular , Animais , Hipóxia Celular , Linhagem Celular , Modelos Animais de Doenças , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestrutura , Fibrose , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/ultraestrutura , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Neovascularização Fisiológica , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Recuperação de Função Fisiológica
16.
CNS Drugs ; 34(6): 559-577, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32472450

RESUMO

There is escalating interest in cell-based therapies to restore lost dopamine inputs in Parkinson's disease. This is based upon the rationale that implanting dopamine progenitors into the striatum can potentially improve dopamine-responsive motor symptoms. A rich body of data describing clinical trials of previous cell transplantation exists. These have included multiple cell sources for transplantation including allogeneic (human embryonic mesencephalic tissue, retinal pigment epithelial cells) and autologous (carotid body, adrenal medullary tissue) cells, as well as xenotransplantation. However, there are multiple limitations related to these cell sources, including availability of adequate numbers of cells for transplant, heterogeneity within cells transplanted, imprecisely defined mechanisms of action, and poor cell survival after transplantation in some cases. Nonetheless, evidence has accrued from a subset of trials to support the rationale for such a regenerative approach. Recent rapid advances in stem cell technology may now overcome these prior limitations. For example, dopamine neuron precursor cells for transplant can be generated from induced pluripotent cells and human embryonic stem cells. The benefits of these innovative approaches include: the possibility of scalability; a high degree of quality control; and improved understanding of mechanisms of action with rigorous preclinical testing. In this review, we focus on the potential for cell-based therapies in Parkinson's disease to restore the function of dopaminergic neurons, we critically review previous attempts to harness such strategies, we discuss potential benefits and predicted limitations, and we address how previous roadblocks may be overcome to bring a cell-based approach to the clinic.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Neurônios Dopaminérgicos/metabolismo , Doença de Parkinson/terapia , Animais , Neurônios Dopaminérgicos/citologia , Células-Tronco Embrionárias Humanas/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Doença de Parkinson/fisiopatologia
17.
Stem Cell Reports ; 14(5): 876-891, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32302555

RESUMO

Huntington disease (HD) is an inherited late-onset neurological disorder characterized by progressive neuronal loss and disruption of cortical and basal ganglia circuits. Cell replacement using human embryonic stem cells may offer the opportunity to repair the damaged circuits and significantly ameliorate disease conditions. Here, we showed that in-vitro-differentiated human striatal progenitors undergo maturation and integrate into host circuits upon intra-striatal transplantation in a rat model of HD. By combining graft-specific immunohistochemistry, rabies virus-mediated synaptic tracing, and ex vivo electrophysiology, we showed that grafts can extend projections to the appropriate target structures, including the globus pallidus, the subthalamic nucleus, and the substantia nigra, and receive synaptic contact from both host and graft cells with 6.6 ± 1.6 inputs cell per transplanted neuron. We have also shown that transplants elicited a significant improvement in sensory-motor tasks up to 2 months post-transplant further supporting the therapeutic potential of this approach.


Assuntos
Corpo Estriado/citologia , Células-Tronco Embrionárias Humanas/transplante , Doença de Huntington/terapia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Animais , Células Cultivadas , Corpo Estriado/fisiologia , Células-Tronco Embrionárias Humanas/citologia , Humanos , Locomoção , Masculino , Células-Tronco Neurais/citologia , Neurogênese , Ratos , Regeneração , Sensação , Substância Negra/citologia , Substância Negra/fisiologia , Núcleo Subtalâmico/citologia , Núcleo Subtalâmico/fisiologia , Sinapses/metabolismo , Sinapses/fisiologia
18.
Cardiovasc Res ; 116(3): 545-553, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31287499

RESUMO

AIMS: Cardiovascular diseases caused by loss of functional cardiomyocytes (CMs) are a major cause of mortality and morbidity worldwide due in part to the low regenerative capacity of the adult human heart. Human pluripotent stem cell (hPSC)-derived cardiovascular progenitor cells (CPCs) are a potential cell source for cardiac repair. The aim of this study was to examine the impact of extensive remuscularization and coincident revascularization on cardiac remodelling and function in a mouse model of myocardial infarction (MI) by transplanting doxycycline (DOX)-inducible (Tet-On-MYC) hPSC-derived CPCs in vivo and inducing proliferation and cardiovascular differentiation in a drug-regulated manner. METHODS AND RESULTS: CPCs were injected firstly at a non-cardiac site in Matrigel suspension under the skin of immunocompromised mice to assess their commitment to the cardiovascular lineage and ability to self-renew or differentiate in vivo when instructed by systemically delivered factors including DOX and basic fibroblast growth factor (bFGF). CPCs in Matrigel were then injected intra-myocardially in mice subjected to MI to assess whether expandable CPCs could mediate cardiac repair. Transplanted CPCs expanded robustly both subcutis and in the myocardium using the same DOX/growth factor inducing regime. Upon withdrawal of these cell-renewal factors, CPCs differentiated with high efficiency at both sites into the major cardiac lineages including CMs, endothelial cells, and smooth muscle cells. After MI, engraftment of CPCs in the heart significantly reduced fibrosis in the infarcted area and prevented left ventricular remodelling, although cardiac function determined by magnetic resonance imaging was unaltered. CONCLUSION: Replacement of large areas of muscle may be required to regenerate the heart of patients following MI. Our human/mouse model demonstrated that proliferating hPSC-CPCs could reduce infarct size and fibrosis resulting in formation of large grafts. Importantly, the results suggested that expanding transplanted cells in situ at the progenitor stage maybe be an effective alternative causing less tissue damage than injection of very large numbers of CMs.


Assuntos
Diferenciação Celular , Proliferação de Células , Células-Tronco Embrionárias Humanas/transplante , Infarto do Miocárdio/cirurgia , Miocárdio/patologia , Miócitos Cardíacos/transplante , Células-Tronco Pluripotentes/transplante , Regeneração , Função Ventricular Esquerda , Animais , Linhagem da Célula , Células Cultivadas , Modelos Animais de Doenças , Fibrose , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Células-Tronco Pluripotentes/metabolismo , Recuperação de Função Fisiológica , Remodelação Ventricular
19.
Neurotherapeutics ; 17(2): 606-608, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-30924090

RESUMO

We examine the ethics of using induced pluripotent stem cells (iPSCs) in cell transplantation treatment of neurologic diseases and the essential types of ethical benchmarks required in clinical trials in neurology using iPSCs, including embryonic pluripotent stem cells. We focus on two issues: (1) comparison and (2) criticism of the two types of neuro-hype (neuro-purism and neuro-essentialism). In order to ensure that the dialog on ethical benchmarks continues to develop in a manner that promotes trust with society and research subjects, concerns about the clinical use of pluripotent stem cells (particularly iPSCs) in neurology must be at the forefront of any ethics discussion.


Assuntos
Benchmarking , Células-Tronco Embrionárias Humanas/transplante , Células-Tronco Pluripotentes Induzidas/transplante , Transplante de Células-Tronco/ética , Humanos
20.
Adv Exp Med Biol ; 1185: 569-574, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31884672

RESUMO

The goal of this study was to quantitatively assess retinal thickness using spectral domain optical coherence tomography (SD-OCT) after subretinal implantation of human embryonic stem cell-derived retinal pigment epithelium in a porcine model. The implant is called CPCB-RPE1 for the California Project to Cure Blindness-Retinal Pigment Epithelium 1. Data were derived from previous experiments on 14 minipigs that received either subretinal implantation of CPCB-RPE1 (n = 11) or subretinal bleb formation alone (sham; n = 3) using previously described methods and procedures (Brant Fernandes et al. Ophthalmic Surg Lasers Imaging Retina 47:342-51, 2016; Martynova et al. (2016) Koss et al. Graefes Arch Clin Exp Ophthalmol 254:1553-65, 2016; Hu et al. Ophthalmic Res 48:186-91, 2016; Martynova et al. ARVO Abstract 2016. SD-OCT retinal thickness (RT) and sublayer thickness over the implant were compared with topographically similar preimplantation regions as described previously Martynova et al. ARVO Abstract 2016. Imaging results were compared to postmortem histology using hematoxylin-eosin staining. RT overlying the CPCB-RPE1 postimplantation was not significantly different from preimplantation (308 ± 72 µm vs 292 ± 41 µm; p = 0.44). RT was not significantly different before and after implantation in any retinal sublayer at 1 month. Histology demonstrated grossly normal retinal anatomy as well as photoreceptor interdigitation with RPE.


Assuntos
Células-Tronco Embrionárias Humanas/transplante , Retina/diagnóstico por imagem , Epitélio Pigmentado da Retina/citologia , Tomografia de Coerência Óptica , Animais , California , Humanos , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA