Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.301
Filter
1.
Neurotherapeutics ; 21(5): e00436, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39353832

ABSTRACT

A breadth of preclinical studies now support the rationale of pluripotent stem cell-derived cell replacement therapies to alleviate motor symptoms in Parkinsonian patients. Replacement of the primary dysfunctional cell population in the disease, i.e. the A9 dopaminergic neurons, is the major focus of these therapies. To achieve this, most therapeutical approaches involve grafting single-cell suspensions of DA progenitors. However, most cells die during the transplantation process, as cells face anoïkis. One potential solution to address this challenge is to graft solid preparations, i.e. adopting a 3D format. Cryopreserving such a format remains a major hurdle and is not exempt from causing delays in the time to effect, as observed with cryopreserved single-cell DA progenitors. Here, we used a high-throughput cell-encapsulation technology coupled with bioreactors to provide a 3D culture environment enabling the directed differentiation of hiPSCs into neural microtissues. The proper patterning of these neural microtissues into a midbrain identity was confirmed using orthogonal methods, including qPCR, RNAseq, flow cytometry and immunofluorescent microscopy. The efficacy of the neural microtissues was demonstrated in a dose-dependent manner using a Parkinsonian rat model. The survival of the cells was confirmed by post-mortem histological analysis, characterised by the presence of human dopaminergic neurons projecting into the host striatum. The work reported here is the first bioproduction of a cell therapy for Parkinson's disease in a scalable bioreactor, leading to a full behavioural recovery 16 weeks after transplantation using cryopreserved 3D format.


Subject(s)
Bioreactors , Dopaminergic Neurons , Induced Pluripotent Stem Cells , Animals , Dopaminergic Neurons/transplantation , Dopaminergic Neurons/physiology , Rats , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/physiology , Cell Differentiation/physiology , Male , Humans , Rats, Sprague-Dawley , Parkinson Disease/therapy , Disease Models, Animal
2.
Med Sci (Paris) ; 40(8-9): 643-652, 2024.
Article in French | MEDLINE | ID: mdl-39303116

ABSTRACT

Rare genetic diseases with neurodevelopmental disorders (NDDs) encompass several heterogeneous conditions (autism spectrum disorder (ASD), intellectual disability (ID), attention deficit hyperactivity disorder (ADHD), specific learning disorder (SLD), among others). Currently, few treatments are available for these patients. The difficulty in accessing human brain samples and the discrepancies between human and animal models highlight the need for new research approaches. One promising approach is the use of the cerebral organoids. These 3D, self-organized structures, generated from induced pluripotent stem cells (iPSCs), enable the reproduction of the stages of human brain development, from the proliferation of neural stem cells to their differentiation into neurons, oligodentrocytes, and astrocytes. Cerebral organoids hold great promise in understanding brain development and in the search for treatments.


Title: Des organoïdes cérébraux pour la compréhension et la thérapie des maladies génétiques rares avec troubles neurodéveloppementaux. Abstract: Les maladies génétiques associées à des troubles neurodéveloppementaux (TND) regroupent plusieurs maladies pour lesquelles peu de traitements sont proposés. L'impossibilité d'accéder à des échantillons de cerveaux humains pour des études ex vivo, et les divergences entre l'homme et les modèles animaux rendent nécessaires de nouvelles approches de recherche. L'organoïde cérébral, une structure en trois dimensions, auto-organisée, et générée à partir de cellules souches pluripotentes induites, permet de reproduire les étapes de développement du cerveau humain, de la prolifération des cellules souches neurales à leur différenciation en neurones, en oligodendrocytes, ou en astrocytes. L'intérêt de ce modèle est désormais prouvé pour la compréhension du développement cérébral et pour la recherche de traitements. Après une présentation des cellules souches pluripotentes induites et des organoïdes, nous exposerons comment cette technique est actuellement déployée, en particulier pour étudier les mécanismes physiopathologiques résultant de variations génétiques pathogènes de gènes candidats de TND.


Subject(s)
Neurodevelopmental Disorders , Organoids , Rare Diseases , Humans , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/therapy , Neurodevelopmental Disorders/pathology , Rare Diseases/genetics , Rare Diseases/therapy , Animals , Induced Pluripotent Stem Cells/transplantation , Brain/pathology , Genetic Diseases, Inborn/therapy , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Cell Differentiation/genetics
3.
Nat Cardiovasc Res ; 3(2): 145-165, 2024 02.
Article in English | MEDLINE | ID: mdl-39196193

ABSTRACT

Preclinical data have confirmed that human pluripotent stem cell-derived cardiomyocytes (PSC-CMs) can remuscularize the injured or diseased heart, with several clinical trials now in planning or recruitment stages. However, because ventricular arrhythmias represent a complication following engraftment of intramyocardially injected PSC-CMs, it is necessary to provide treatment strategies to control or prevent engraftment arrhythmias (EAs). Here, we show in a porcine model of myocardial infarction and PSC-CM transplantation that EAs are mechanistically linked to cellular heterogeneity in the input PSC-CM and resultant graft. Specifically, we identify atrial and pacemaker-like cardiomyocytes as culprit arrhythmogenic subpopulations. Two unique surface marker signatures, signal regulatory protein α (SIRPA)+CD90-CD200+ and SIRPA+CD90-CD200-, identify arrhythmogenic and non-arrhythmogenic cardiomyocytes, respectively. Our data suggest that modifications to current PSC-CM-production and/or PSC-CM-selection protocols could potentially prevent EAs. We further show that pharmacologic and interventional anti-arrhythmic strategies can control and potentially abolish these arrhythmias.


Subject(s)
Arrhythmias, Cardiac , Myocytes, Cardiac , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/transplantation , Animals , Arrhythmias, Cardiac/therapy , Humans , Disease Models, Animal , Myocardial Infarction/therapy , Swine , Cells, Cultured , Cell Differentiation , Induced Pluripotent Stem Cells/transplantation , Action Potentials/physiology , Action Potentials/drug effects , Phenotype , Biomarkers/metabolism , Pluripotent Stem Cells/transplantation , Stem Cell Transplantation/methods , Anti-Arrhythmia Agents/therapeutic use , Anti-Arrhythmia Agents/pharmacology , Heart Rate/physiology
4.
Nat Commun ; 15(1): 5834, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992003

ABSTRACT

We present Dystrophic Epidermolysis Bullosa Cell Therapy (DEBCT), a scalable platform producing autologous organotypic iPS cell-derived induced skin composite (iSC) grafts for definitive treatment. Clinical-grade manufacturing integrates CRISPR-mediated genetic correction with reprogramming into one step, accelerating derivation of COL7A1-edited iPS cells from patients. Differentiation into epidermal, dermal and melanocyte progenitors is followed by CD49f-enrichment, minimizing maturation heterogeneity. Mouse xenografting of iSCs from four patients with different mutations demonstrates disease modifying activity at 1 month. Next-generation sequencing, biodistribution and tumorigenicity assays establish a favorable safety profile at 1-9 months. Single cell transcriptomics reveals that iSCs are composed of the major skin cell lineages and include prominent holoclone stem cell-like signatures of keratinocytes, and the recently described Gibbin-dependent signature of fibroblasts. The latter correlates with enhanced graftability of iSCs. In conclusion, DEBCT overcomes manufacturing and safety roadblocks and establishes a reproducible, safe, and cGMP-compatible therapeutic approach to heal lesions of DEB patients.


Subject(s)
Cell- and Tissue-Based Therapy , Collagen Type VII , Epidermolysis Bullosa Dystrophica , Induced Pluripotent Stem Cells , Humans , Epidermolysis Bullosa Dystrophica/therapy , Epidermolysis Bullosa Dystrophica/genetics , Animals , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Mice , Collagen Type VII/genetics , Collagen Type VII/metabolism , Cell- and Tissue-Based Therapy/methods , Fibroblasts/metabolism , Cell Differentiation , Keratinocytes/metabolism , Keratinocytes/transplantation , Skin/metabolism , Transplantation, Autologous , Male , Mutation , Female , Skin Transplantation/methods , Gene Editing/methods , CRISPR-Cas Systems
5.
Cell Mol Life Sci ; 81(1): 318, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073571

ABSTRACT

Nerve regeneration and circuit reconstruction remain a challenge following spinal cord injury (SCI). Corticospinal pyramidal neurons possess strong axon projection ability. In this study, human induced pluripotent stem cells (iPSCs) were differentiated into pyramidal neuronal precursors (PNPs) by addition of small molecule dorsomorphin into the culture. iPSC-derived PNPs were transplanted acutely into a rat contusion SCI model on the same day of injury. Following engraftment, the SCI rats showed significantly improved motor functions compared with vehicle control group as revealed by behavioral tests. Eight weeks following engraftment, the PNPs matured into corticospinal pyramidal neurons and extended axons into distant host spinal cord tissues, mostly in a caudal direction. Host neurons rostral to the lesion site also grew axons into the graft. Possible synaptic connections as a bridging relay may have been formed between host and graft-derived neurons, as indicated by pre- and post-synaptic marker staining and the regulation of chemogenetic regulatory systems. PNP graft showed an anti-inflammatory effect at the injury site and could bias microglia/macrophages towards a M2 phenotype. In addition, PNP graft was safe and no tumor formation was detected after transplantation into immunodeficient mice and SCI rats. The potential to reconstruct a neuronal relay circuitry across the lesion site and to modulate the microenvironment in SCI makes PNPs a promising cellular candidate for treatment of SCI.


Subject(s)
Cell Differentiation , Disease Models, Animal , Induced Pluripotent Stem Cells , Spinal Cord Injuries , Animals , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Mice , Neural Stem Cells/transplantation , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Nerve Regeneration , Axons/metabolism
6.
Exp Neurol ; 379: 114848, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38857749

ABSTRACT

The establishment of reliable human brain models is pivotal for elucidating specific disease mechanisms and facilitating the discovery of novel therapeutic strategies for human brain disorders. Human induced pluripotent stem cell (iPSC) exhibit remarkable self-renewal capabilities and can differentiate into specialized cell types. This makes them a valuable cell source for xenogeneic or allogeneic transplantation. Human-mouse chimeric brain models constructed from iPSC-derived brain cells have emerged as valuable tools for modeling human brain diseases and exploring potential therapeutic strategies for brain disorders. Moreover, the integration and functionality of grafted stem cells has been effectively assessed using these models. Therefore, this review provides a comprehensive overview of recent progress in differentiating human iPSC into various highly specialized types of brain cells. This review evaluates the characteristics and functions of the human-mouse chimeric brain model. We highlight its potential roles in brain function and its ability to reconstruct neural circuitry in vivo. Additionally, we elucidate factors that influence the integration and differentiation of human iPSC-derived brain cells in vivo. This review further sought to provide suitable research models for cell transplantation therapy. These research models provide new insights into neuropsychiatric disorders, infectious diseases, and brain injuries, thereby advancing related clinical and academic research.


Subject(s)
Brain , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/physiology , Animals , Brain/cytology , Mice , Cell Differentiation/physiology , Chimera , Disease Models, Animal , Brain Diseases/therapy
7.
Stem Cells ; 42(9): 781-790, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38902932

ABSTRACT

The motor symptoms of Parkinson's disease (PD) are caused by the progressive loss of dopamine neurons from the substantia nigra. There are currently no treatments that can slow or reverse the neurodegeneration. To restore the lost neurons, international groups have initiated clinical trials using human embryonic or induced pluripotent stem cells (PSCs) to derive dopamine neuron precursors that are used as transplants to replace the lost neurons. Proof-of-principle experiments in the 1980s and 1990s showed that grafts of fetal ventral mesencephalon, which contains the precursors of the substantial nigra, could, under rare circumstances, reverse symptoms of the disease. Improvements in PSC technology and genomics have inspired researchers to design clinical trials using PSC-derived dopamine neuron precursors as cell replacement therapy for PD. We focus here on 4 such first-in-human clinical trials that have begun in the US, Europe, and Japan. We provide an overview of the sources of PSCs and the methods used to generate cells for transplantation. We discuss pros and cons of strategies for allogeneic, immune-matched, and autologous approaches and novel methods for overcoming rejection by the immune system. We consider challenges for safety and efficacy of the cells for durable engraftment, focusing on the genomics-based quality control methods to assure that the cells will not become cancerous. Finally, since clinical trials like these have never been undertaken before, we comment on the value of cooperation among rivals to contribute to advancements that will finally provide relief for the millions suffering from the symptoms of PD.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/therapy , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Dopaminergic Neurons/transplantation , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/cytology , Cell- and Tissue-Based Therapy/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Stem Cell Transplantation/methods , Animals , Cell Differentiation
8.
Neuron ; 112(16): 2686-2707.e8, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-38897209

ABSTRACT

Microglia replacement strategies are increasingly being considered for the treatment of primary microgliopathies like adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). However, available mouse models fail to recapitulate the diverse neuropathologies and reduced microglia numbers observed in patients. In this study, we generated a xenotolerant mouse model lacking the fms-intronic regulatory element (FIRE) enhancer within Csf1r, which develops nearly all the hallmark pathologies associated with ALSP. Remarkably, transplantation of human induced pluripotent stem cell (iPSC)-derived microglial (iMG) progenitors restores a homeostatic microglial signature and prevents the development of axonal spheroids, white matter abnormalities, reactive astrocytosis, and brain calcifications. Furthermore, transplantation of CRISPR-corrected ALSP-patient-derived iMG reverses pre-existing spheroids, astrogliosis, and calcification pathologies. Together with the accompanying study by Munro and colleagues, our results demonstrate the utility of FIRE mice to model ALSP and provide compelling evidence that iMG transplantation could offer a promising new therapeutic strategy for ALSP and perhaps other microglia-associated neurological disorders.


Subject(s)
Disease Models, Animal , Induced Pluripotent Stem Cells , Leukoencephalopathies , Microglia , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Animals , Microglia/metabolism , Leukoencephalopathies/genetics , Humans , Mice , Induced Pluripotent Stem Cells/transplantation , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Gliosis , Receptor, Macrophage Colony-Stimulating Factor
9.
Nat Med ; 30(6): 1556-1558, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38778211

ABSTRACT

The first completed clinical trial of induced pluripotent stem cell (iPS cell)-derived cells was conducted in 15 participants with steroid-resistant acute graft-versus-host disease. After intravenous infusion of mesenchymal stromal cells (CYP-001 derived from a clone of human iPS cells), we reported the safety, tolerability and efficacy within the primary evaluation period at day 100. We now report results at the 2-year follow-up: 9 of 15 (60%) participants survived, which compares favorably with previously reported outcomes in studies of steroid-resistant acute graft-versus-host disease. Causes of death were complications commonly observed in recipients of allogeneic hematopoietic stem cell transplantation, and not considered by the investigators to be related to CYP-001 treatment. There were no serious adverse events, tumors or other safety concerns related to CYP-001. In conclusion, systemic delivery of iPS cell-derived cells was safe and well tolerated over 2 years of follow-up, with sustained outcomes up to 2 years after the first infusion. ClinicalTrials.gov registration: NCT02923375 .


Subject(s)
Graft vs Host Disease , Induced Pluripotent Stem Cells , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Induced Pluripotent Stem Cells/transplantation , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/immunology , Male , Adult , Female , Middle Aged , Steroids/therapeutic use , Treatment Outcome , Hematopoietic Stem Cell Transplantation/adverse effects , Follow-Up Studies , Acute Disease , Drug Resistance , Young Adult , Transplantation, Homologous/adverse effects
10.
J Clin Invest ; 134(12)2024 May 07.
Article in English | MEDLINE | ID: mdl-38713532

ABSTRACT

Satellite cells, the stem cells of skeletal muscle tissue, hold a remarkable regeneration capacity and therapeutic potential in regenerative medicine. However, low satellite cell yield from autologous or donor-derived muscles hinders the adoption of satellite cell transplantation for the treatment of muscle diseases, including Duchenne muscular dystrophy (DMD). To address this limitation, here we investigated whether satellite cells can be derived in allogeneic or xenogeneic animal hosts. First, injection of CRISPR/Cas9-corrected Dmdmdx mouse induced pluripotent stem cells (iPSCs) into mouse blastocysts carrying an ablation system of host satellite cells gave rise to intraspecies chimeras exclusively carrying iPSC-derived satellite cells. Furthermore, injection of genetically corrected DMD iPSCs into rat blastocysts resulted in the formation of interspecies rat-mouse chimeras harboring mouse satellite cells. Notably, iPSC-derived satellite cells or derivative myoblasts produced in intraspecies or interspecies chimeras restored dystrophin expression in DMD mice following intramuscular transplantation and contributed to the satellite cell pool. Collectively, this study demonstrates the feasibility of producing therapeutically competent stem cells across divergent animal species, raising the possibility of generating human muscle stem cells in large animals for regenerative medicine purposes.


Subject(s)
Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Satellite Cells, Skeletal Muscle , Animals , Mice , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/genetics , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Rats , Satellite Cells, Skeletal Muscle/transplantation , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Stem Cell Transplantation , Humans , Dystrophin/genetics , Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Mice, Inbred mdx , Heterografts , Transplantation, Heterologous , Injections, Intramuscular , Transplantation, Homologous
11.
J Cereb Blood Flow Metab ; 44(7): 1246-1249, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38629577

ABSTRACT

Cell therapy and regenerative medicine have made remarkable progress in treating neurodegenerative disorders. Induced pluripotent stem cells (iPSCs) offer a promising source for cell replacement therapies, but their practical application faces challenges due to poor survival and integration after transplantation. Park et al. propose a novel therapeutic strategy involving the co-transplantation of regulatory T cells (Tregs) and iPSC-derived dopamine neurons. This combined approach enhances the survival of transplanted cells and protects against neuroinflammation-induced damage. In PD animal models, the co-transplantation approach significantly suppressed the host immune response, resulting in improved behavioral recovery. Additionally, Tregs demonstrate acute neuroprotection and contribute to delayed neuro-restoration in ischemic stroke. This combined approach of cell therapy with immunomodulation offers a promising avenue for advancing our understanding of neurological diseases and promoting the development of novel treatments.


Subject(s)
Brain Diseases , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , Humans , Animals , Brain Diseases/therapy , Brain Diseases/immunology , Induced Pluripotent Stem Cells/transplantation , Cell- and Tissue-Based Therapy/methods , Transplantation, Autologous/methods
12.
Circulation ; 150(8): 611-621, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38666382

ABSTRACT

BACKGROUND: The clinical application of human induced pluripotent stem cell-derived cardiomyocytes (CMs) for cardiac repair commenced with the epicardial delivery of engineered cardiac tissue; however, the feasibility of the direct delivery of human induced pluripotent stem cell-derived CMs into the cardiac muscle layer, which has reportedly induced electrical integration, is unclear because of concerns about poor engraftment of CMs and posttransplant arrhythmias. Thus, in this study, we prepared purified human induced pluripotent stem cell-derived cardiac spheroids (hiPSC-CSs) and investigated whether their direct injection could regenerate infarcted nonhuman primate hearts. METHODS: We performed 2 separate experiments to explore the appropriate number of human induced pluripotent stem cell-derived CMs. In the first experiment, 10 cynomolgus monkeys were subjected to myocardial infarction 2 weeks before transplantation and were designated as recipients of hiPSC-CSs containing 2×107 CMs or the vehicle. The animals were euthanized 12 weeks after transplantation for histological analysis, and cardiac function and arrhythmia were monitored during the observational period. In the second study, we repeated the equivalent transplantation study using more CMs (6×107 CMs). RESULTS: Recipients of hiPSC-CSs containing 2×107 CMs showed limited CM grafts and transient increases in fractional shortening compared with those of the vehicle (fractional shortening at 4 weeks after transplantation [mean ± SD]: 26.2±2.1%; 19.3±1.8%; P<0.05), with a low incidence of posttransplant arrhythmia. Transplantation of increased dose of CMs resulted in significantly greater engraftment and long-term contractile benefits (fractional shortening at 12 weeks after transplantation: 22.5±1.0%; 16.6±1.1%; P<0.01, left ventricular ejection fraction at 12 weeks after transplantation: 49.0±1.4%; 36.3±2.9%; P<0.01). The incidence of posttransplant arrhythmia slightly increased in recipients of hiPSC-CSs containing 6×107 CMs. CONCLUSIONS: We demonstrated that direct injection of hiPSC-CSs restores the contractile functions of injured primate hearts with an acceptable risk of posttransplant arrhythmia. Although the mechanism for the functional benefits is not fully elucidated, these findings provide a strong rationale for conducting clinical trials using the equivalent CM products.


Subject(s)
Induced Pluripotent Stem Cells , Macaca fascicularis , Myocardial Infarction , Myocytes, Cardiac , Spheroids, Cellular , Animals , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/cytology , Humans , Myocytes, Cardiac/transplantation , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Spheroids, Cellular/transplantation , Regeneration , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/pathology , Male , Stem Cell Transplantation/methods , Disease Models, Animal
13.
J Neural Eng ; 21(2)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38479026

ABSTRACT

Objective.Although human induced pluripotent stem cell (iPSC)-derived cell replacement for Parkinson's disease has considerable reparative potential, its full therapeutic benefit is limited by poor graft survival and dopaminergic maturation. Injectable biomaterial scaffolds, such as collagen hydrogels, have the potential to address these issues via a plethora of supportive benefits including acting as a structural scaffold for cell adherence, shielding from the host immune response and providing a reservoir of neurotrophic factors to aid survival and differentiation. Thus, the aim of this study was to determine if a neurotrophin-enriched collagen hydrogel could improve the survival and maturation of iPSC-derived dopaminergic progenitors (iPSC-DAPs) after transplantation into the rat parkinsonian brain.Approach.Human iPSC-DAPs were transplanted into the 6-hydroxydopamine-lesioned striatum either alone, with the neurotrophins GDNF and BDNF, in an unloaded collagen hydrogel, or in a neurotrophin-loaded collagen hydrogel.Post-mortem, human nuclear immunostaining was used to identify surviving iPSC-DAPs while tyrosine hydroxylase immunostaining was used to identify iPSC-DAPs that had differentiated into mature dopaminergic neurons.Main results.We found that iPSC-DAPs transplanted in the neurotrophin-enriched collagen hydrogel survived and matured significantly better than cells implanted without the biomaterial (8 fold improvement in survival and 16 fold improvement in dopaminergic differentiation). This study shows that transplantation of human iPSC-DAPs in a neurotrophin-enriched collagen hydrogel improves graft survival and maturation in the parkinsonian rat brain.Significance.The data strongly supports further investigation of supportive hydrogels for improving the outcome of iPSC-derived brain repair in Parkinson's disease.


Subject(s)
Induced Pluripotent Stem Cells , Parkinson Disease , Rats , Animals , Humans , Nerve Growth Factors/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Hydrogels/chemistry , Parkinson Disease/therapy , Brain/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/transplantation , Biocompatible Materials , Collagen , Cell Differentiation
14.
Transplantation ; 108(10): e282-e291, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38416452

ABSTRACT

Chronic lung diseases are debilitating illnesses ranking among the top causes of death globally. Currently, clinically available therapeutic options capable of curing chronic lung diseases are limited to lung transplantation, which is hindered by donor organ shortage. This highlights the urgent need for alternative strategies to repair damaged lung tissues. Stem cell transplantation has emerged as a promising avenue for regenerative treatment of the lung, which involves delivery of healthy lung epithelial progenitor cells that subsequently engraft in the injured tissue and further differentiate to reconstitute the functional respiratory epithelium. These transplanted progenitor cells possess the remarkable ability to self-renew, thereby offering the potential for sustained long-term treatment effects. Notably, the transplantation of basal cells, the airway stem cells, holds the promise for rehabilitating airway injuries resulting from environmental factors or genetic conditions such as cystic fibrosis. Similarly, for diseases affecting the alveoli, alveolar type II cells have garnered interest as a viable alveolar stem cell source for restoring the lung parenchyma from genetic or environmentally induced dysfunctions. Expanding upon these advancements, the use of induced pluripotent stem cells to derive lung progenitor cells for transplantation offers advantages such as scalability and patient specificity. In this review, we comprehensively explore the progress made in lung stem cell transplantation, providing insights into the current state of the field and its future prospects.


Subject(s)
Lung Diseases , Lung , Regeneration , Regenerative Medicine , Stem Cell Transplantation , Humans , Lung Diseases/therapy , Lung Diseases/surgery , Stem Cell Transplantation/methods , Lung/physiopathology , Regenerative Medicine/methods , Animals , Cell Differentiation , Induced Pluripotent Stem Cells/transplantation , Stem Cells/cytology
15.
Stem Cell Reports ; 19(3): 331-342, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38335965

ABSTRACT

Several retinal degenerations affect the human central retina, which is primarily comprised of cones and is essential for high acuity and color vision. Transplanting cone photoreceptors is a promising strategy to replace degenerated cones in this region. Although this approach has been investigated in a handful of animal models, commonly used rodent models lack a cone-rich region and larger models can be expensive and inaccessible, impeding the translation of therapies. Here, we transplanted dissociated GFP-expressing photoreceptors from retinal organoids differentiated from human induced pluripotent stem cells into the subretinal space of damaged and undamaged cone-dominant 13-lined ground squirrel eyes. Transplanted cell survival was documented via noninvasive high-resolution imaging and immunohistochemistry to confirm the presence of human donor photoreceptors for up to 4 months posttransplantation. These results demonstrate the utility of a cone-dominant rodent model for advancing the clinical translation of cell replacement therapies.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Degeneration , Animals , Humans , Retinal Cone Photoreceptor Cells/transplantation , Induced Pluripotent Stem Cells/transplantation , Retina , Retinal Degeneration/therapy , Sciuridae
16.
Stem Cell Res Ther ; 15(1): 35, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38321505

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) is a devastating disease that causes extensive damage to oligodendrocytes and neurons leading to demyelination and axonal degeneration. In this study, we co-transplanted cell grafts containing oligodendrocyte progenitor cells (OPCs) derived from human-induced pluripotent stem cells (iPSCs) combined with human umbilical vein endothelial cells (HUVECs), which were reported to promote OPCs survival and migration, into rat contusion models to promote functional recovery after SCI. METHODS: OPCs were derived from iPSCs and identified by immunofluorescence at different time points. Functional assays in vitro were performed to evaluate the effect of HUVECs on the proliferation, migration, and survival of OPCs by co-culture and migration assay, as well as on the neuronal axonal growth. A combination of OPCs and HUVECs was transplanted into the rat contusive model. Upon 8 weeks, immunofluorescence staining was performed to test the safety of transplanted cells and to observe the neuronal repairment, myelination, and neural circuit reconstruction at the injured area; also, the functional recovery was assessed by Basso, Beattie, and Bresnahan open-field scale, Ladder climb, SEP, and MEP. Furthermore, the effect of HUVECs on grafts was also determined in vivo. RESULTS: Data showed that HUVECs promote the proliferation, migration, and survival of OPCs both in vitro and in vivo. Furthermore, 8 weeks upon engraftment, the rats with OPCs and HUVECs co-transplantation noticeably facilitated remyelination, enhanced functional connection between the grafts and the host and promoted functional recovery. In addition, compared with the OPCs-alone transplantation, the co-transplantation generated more sensory neurons at the lesion border and significantly improved the sensory functional recovery. CONCLUSIONS: Our study demonstrates that transplantation of OPCs combined with HUVECs significantly enhances both motor and sensory functional recovery after SCI. No significance was observed between OPCs combined with HUVECs group and OPCs-alone group in motor function recovery, while the sensory function recovery was significantly promoted in OPCs combined with HUVECs groups compared with the other two groups. These findings provide novel insights into the field of SCI research.


Subject(s)
Induced Pluripotent Stem Cells , Oligodendrocyte Precursor Cells , Spinal Cord Injuries , Rats , Humans , Animals , Oligodendrocyte Precursor Cells/pathology , Oligodendrocyte Precursor Cells/transplantation , Human Umbilical Vein Endothelial Cells , Recovery of Function , Induced Pluripotent Stem Cells/transplantation , Spinal Cord Injuries/pathology , Oligodendroglia , Spinal Cord/pathology , Cell Differentiation/physiology
17.
Exp Neurol ; 374: 114694, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38272159

ABSTRACT

Parkinson's disease (PD) is a relentlessly progressive and currently incurable neurodegenerative disease with significant unmet medical needs. Since PD stems from the degeneration of midbrain dopaminergic (DA) neurons in a defined brain location, PD patients are considered optimal candidates for cell replacement therapy. Clinical trials for cell transplantation in PD are beginning to re-emerge worldwide with a new focus on induced pluripotent stem cells (iPSCs) as a source of DA neurons since they can be derived from adult somatic cells and produced in large quantities under current good manufacturing practices. However, for this therapeutic strategy to be realized as a viable clinical option, fundamental translational challenges need to be addressed including the manufacturing process, purity and efficacy of the cells, the method of delivery, the extent of host reinnervation and the impact of patient-centered adjunctive interventions. In this study we report on the impact of physical and cognitive training (PCT) on functional recovery in the nonhuman primate (NHP) model of PD after cell transplantation. We observed that at 6 months post-transplant, the PCT group returned to normal baseline in their daily activity measured by actigraphy, significantly improved in their sensorimotor and cognitive tasks, and showed enhanced synapse formation between grafted cells and host cells. We also describe a robust, simple, efficient, scalable, and cost-effective manufacturing process of engraftable DA neurons derived from iPSCs. This study suggests that integrating PCT with cell transplantation therapy could promote optimal graft functional integration and better outcome for patients with PD.


Subject(s)
Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Parkinson Disease , Adult , Animals , Humans , Dopaminergic Neurons/physiology , Induced Pluripotent Stem Cells/transplantation , Callithrix , Cognitive Training , Parkinson Disease/surgery , Stem Cell Transplantation/methods , Cell Differentiation/physiology
18.
Expert Opin Biol Ther ; 23(9): 883-899, 2023.
Article in English | MEDLINE | ID: mdl-37545020

ABSTRACT

INTRODUCTION: Spinal cord injury (SCI) can lead to severe neurological dysfunction. Despite scientific and medical advances, clinically effective regenerative therapies including stem cells are lacking for SCI. AREAS COVERED: This paper discusses translational challenges related to the safe, effective use of stem cells for SCI, with a focus on mesenchymal stem cells (MSCs), neural stem cells (NSCs), Schwann cells (SCs), olfactory ensheathing cells (OECs), oligodendrocyte precursor cells (OPCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs). We discuss approaches to enhance the efficacy of cell-based strategies by i) addressing patient heterogeneity and enhancing patient selection; ii) selecting cell type, cell source, cell developmental stage, and delivery technique; iii) enhancing graft integration and mitigating immune-mediated graft rejection; and iv) ensuring availability of cells. Additionally, we review strategies to optimize outcomes including combinatorial use of rehabilitation and discuss ways to mitigate potential risks of tumor formation associated with stem cell-based strategies. EXPERT OPINION: Basic science research will drive translational advances to develop stem cell-based therapies for SCI. Genetic, serological, and imaging biomarkers may enable individualization of cell-based treatments. Moreover, combinatorial strategies will be required to enhance graft survival, migration and functional integration, to enable precision-based intervention.


Subject(s)
Induced Pluripotent Stem Cells , Neural Stem Cells , Spinal Cord Injuries , Humans , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/methods , Embryonic Stem Cells , Induced Pluripotent Stem Cells/transplantation , Spinal Cord
19.
Biochem Biophys Res Commun ; 674: 190-198, 2023 09 24.
Article in English | MEDLINE | ID: mdl-37532637

ABSTRACT

Cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) are a promising cell source for regenerative medicine and drug discovery. However, the use of animal models for studying human cardiomyocytes derived from hiPSCs in vivo is limited and challenging. Given the shared properties between humans and zebrafish, their ethical advantages over mammalian models, and their immature immune system that is rejection-free against xenografted human cells, zebrafish provide a suitable alternative model for xenograft studies. We microinjected fluorescence-labeled cardiac lineage cells derived from hiPSCs, specifically mesoderm or cardiac mesoderm cells, into the yolk and the area proximal to the outflow tract of the linear heart at 24 hours post-fertilization (hpf). The cells injected into the yolk survived and did not migrate to other tissues. In contrast, the cells injected contiguous with the outflow tract of the linear heart migrated into the pericardial cavity and heart. After 1 day post injection (1 dpi, 22-24 hpi), the injected cells migrated into the pericardial cavity and heart. Importantly, we observed heartbeat-like movements of some injected cells in the zebrafish heart after 1 dpi. These results suggested successful xenografting of hiPSC-derived cardiac lineage cells into the zebrafish embryo heart. Thus, we developed a valuable tool using zebrafish embryos as a model organism for investigating the molecular and cellular mechanisms involved in the grafting process. This is essential in developing cell transplantation-based cardiac therapeutics as well as for drug testing, notably contributing to advancements in the field of cardio-medicine.


Subject(s)
Induced Pluripotent Stem Cells , Zebrafish , Animals , Humans , Induced Pluripotent Stem Cells/transplantation , Cell Differentiation , Transplantation, Heterologous , Heterografts , Myocytes, Cardiac , Mammals
20.
Nature ; 619(7970): 606-615, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37438521

ABSTRACT

The specific loss of midbrain dopamine neurons (mDANs) causes major motor dysfunction in Parkinson's disease, which makes cell replacement a promising therapeutic approach1-4. However, poor survival of grafted mDANs remains an obstacle to successful clinical outcomes5-8. Here we show that the surgical procedure itself (referred to here as 'needle trauma') triggers a profound host response that is characterized by acute neuroinflammation, robust infiltration of peripheral immune cells and brain cell death. When midbrain dopamine (mDA) cells derived from human induced pluripotent stem (iPS) cells were transplanted into the rodent striatum, less than 10% of implanted tyrosine hydroxylase (TH)+ mDANs survived at two weeks after transplantation. By contrast, TH- grafted cells mostly survived. Notably, transplantation of autologous regulatory T (Treg) cells greatly modified the response to needle trauma, suppressing acute neuroinflammation and immune cell infiltration. Furthermore, intra-striatal co-transplantation of Treg cells and human-iPS-cell-derived mDA cells significantly protected grafted mDANs from needle-trauma-associated death and improved therapeutic outcomes in rodent models of Parkinson's disease with 6-hydroxydopamine lesions. Co-transplantation with Treg cells also suppressed the undesirable proliferation of TH- grafted cells, resulting in more compact grafts with a higher proportion and higher absolute numbers of TH+ neurons. Together, these data emphasize the importance of the initial inflammatory response to surgical injury in the differential survival of cellular components of the graft, and suggest that co-transplanting autologous Treg cells effectively reduces the needle-trauma-induced death of mDANs, providing a potential strategy to achieve better clinical outcomes for cell therapy in Parkinson's disease.


Subject(s)
Cell- and Tissue-Based Therapy , Dopaminergic Neurons , Graft Survival , Neuroinflammatory Diseases , Parkinson Disease , T-Lymphocytes, Regulatory , Tyrosine 3-Monooxygenase , Humans , Dopamine/analogs & derivatives , Dopamine/metabolism , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/transplantation , Mesencephalon/pathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/prevention & control , Neuroinflammatory Diseases/therapy , Parkinson Disease/complications , Parkinson Disease/pathology , Parkinson Disease/surgery , Parkinson Disease/therapy , Tyrosine 3-Monooxygenase/deficiency , Tyrosine 3-Monooxygenase/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Cell- and Tissue-Based Therapy/methods , Animals , Mice , Rats , Oxidopamine/metabolism , Graft Survival/immunology , Cell Death , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Neostriatum/metabolism , Time Factors , Cell Proliferation , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL