Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 321
Filtrar
1.
Front Immunol ; 15: 1410784, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39156892

RESUMEN

Background: PaBing-II Formula (PB-II) is a traditional Chinese medicine for treating Parkinson's disease (PD). However, owing to the complexity of PB-II and the difficulty in obtaining human dopaminergic neurons (DAn), the mechanism of action of PB-II in PD treatment remains unclear. The aim of this study was to investigate the mechanisms underlying the therapeutic benefits of PB-II in patients with PD. Methods: hiPSCs derived DAn were treated with H2O2 to construct the DAn oxidative damage model. SwissTargetPrediction was employed to predict the potential targets of the main compounds in serum after PB-II treatment. Metascape was used to analyze the pathways. Sprague-Dawley rats were used to construct the 6-hydroxydopamine (6-OHDA)-induced PD model, and the duration of administration was four weeks. RNA sequencing was used for Transcriptome analysis to find the signal pathways related to neuronal damage. The associated inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA). We identified PB-II as an Nrf2 activator using antioxidant-responsive element luciferase assay in MDA-MB-231 cells. Results: In vitro experiments showed that the treatment of PB-II-treated serum increased the percentage of TH+ cells, decreased inflammation and the apoptosis, reduced cellular reactive oxygen species, and upregulated the expression of Nrf2 and its downstream genes. Pathway analysis of the RNA-seq data of samples before and after the treatment with PB-II-treated serum identified neuron-associated pathways. In vivo experiments demonstrated that PB-II treatment of PD rat model could activate the Nrf2 signaling pathway, protect the midbrain DAn, and improve the symptoms in PD rats. Conclusion: PB-II significantly protects DAn from inflammation and oxidative stress via Nrf2 pathway activation. These findings elucidate the roles of PB-II in PD treatment and demonstrate the application of hiPSC-derived DAn in research of Chinese medicine.


Asunto(s)
Neuronas Dopaminérgicas , Medicamentos Herbarios Chinos , Células Madre Pluripotentes Inducidas , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Ratas Sprague-Dawley , Humanos , Estrés Oxidativo/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Animales , Medicamentos Herbarios Chinos/farmacología , Ratas , Células Madre Pluripotentes Inducidas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Masculino , Transducción de Señal/efectos de los fármacos , Modelos Animales de Enfermedad , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Oxidopamina , Medicina Tradicional China
2.
Atherosclerosis ; 397: 118549, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39141976

RESUMEN

BACKGROUND AND AIMS: The objective of this study was to investigate the efficacy of CRISPR/Cas9-mediated A4GALT suppression in rescuing endothelial dysfunction in Fabry disease (FD) endothelial cells (FD-ECs) derived from human induced pluripotent stem cells (hiPSCs). METHODS: We differentiated hiPSCs (WT (wild-type), WTC-11), GLA-mutant hiPSCs (GLA-KO, CMC-Fb-002), and CRISPR/Cas9-mediated A4GALT-KO hiPSCs (GLA/A4GALT-KO, Fb-002-A4GALT-KO) into ECs and compared FD phenotypes and endothelial dysfunction. We also analyzed the effect of A4GALT suppression on reactive oxygen species (ROS) formation and transcriptome profiles through RNA sequencing. RESULTS: GLA-mutant hiPSC-ECs (GLA-KO and CMC-Fb-002) showed downregulated expression of EC markers and significantly reduced α-GalA expression with increased Gb-3 deposition and intra-lysosomal inclusion bodies. However, CRISPR/Cas9-mediated A4GALT suppression in GLA/A4GALT-KO and Fb-002-A4GALT-KO hiPSC-ECs increased expression levels of EC markers and rescued these FD phenotypes. GLA-mutant hiPSC-ECs failed to form tube-like structure in tube formation assays, showing significantly decreased migration of cells into the scratched wound area. In contrast, A4GALT suppression improved tube formation and cell migration capacity. Western blot analysis revealed that MAPK and AKT phosphorylation levels were downregulated while SOD and catalase were upregulated in GLA-KO hiPSC-ECs. However, suppression of A4GALT restored these protein alterations. RNA sequencing analysis demonstrated significant transcriptome changes in GLA-mutant EC, especially in angiogenesis, cell death, and cellular response to oxidative stress. However, these were effectively restored in GLA/A4GALT-KO hiPSC-ECs. CONCLUSIONS: CRISPR/Cas9-mediated A4GALT suppression rescued FD phenotype and endothelial dysfunction in GLA-mutant hiPSC-ECs, presenting a potential therapeutic approach for FD-vasculopathy.

3.
Mol Ther Methods Clin Dev ; 32(3): 101302, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39185274

RESUMEN

As autologous induced pluripotent stem cell (iPSC) therapy requires a custom-made small-lot cell production line, and the cell production method differs significantly from the existing processes for producing allogeneic iPSC stocks for clinical use. Specifically, mass culture to produce stock is no longer necessary; instead, a series of operations from iPSC production to induction of differentiation of therapeutic cells must be performed continuously. A three-dimensional (3D) culture method using small, closed-cell manufacturing devices is suitable for autologous iPSC therapy. The use of such devices avoids the need to handle many patient-derived specimens in a single clean room; handling of cell cultures in an open system in a cell processing facility increases the risk of infection. In this study, atelocollagen beads were evaluated as a 3D biomaterial to assist 3D culture in the establishment, expansion culture, and induction of differentiation of iPSCs. It was found that iPSCs can be handled in a closed-cell device with the same ease as use of a two-dimensional (2D) culture when laminin-511 is added to the medium. In conclusion, atelocollagen beads enable 3D culture of iPSCs, and the quality of the obtained cells is at the same level as those derived from 2D culture.

4.
J Neurooncol ; 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39196480

RESUMEN

PURPOSE: Glioblastoma (GBM) is the most prevalent, malignant, primary brain tumor in adults, characterized by limited treatment options, frequent relapse, and short survival after diagnosis. Until now, none of the existing therapy and treatment approaches have proven to be an effective cure. The availability of predictive human blood-tumor barrier (BTB) test systems that can mimic in-vivo pathophysiology of GBM would be of great interest in preclinical research. Here, we present the establishment of a new BTB in-vitro test system combining GBM spheroids and BBB models derived from human induced pluripotent stem cells (hiPSCs). METHODS: We co-cultured hiPSC-derived brain capillary endothelial-like cells (iBCECs) with GBM spheroids derived from U87-MG and U373-MG cell lines in a cell culture insert-based format. Spheroids were monitored over 168 hours (h) of culture, characterized for GBM-specific marker expression and treated with standard chemotherapeutics to distinguish inhibitory effects between 2D mono-culture and 3D spheroids. GBM-induced changes on iBCECs barrier integrity were verified via measurement of transendothelial electrical resistance (TEER), immunocytochemical staining of tight junction (TJ) proteins claudin-5 and occludin as well as the glucose transporter-1 (Glut-1). GBM-induced secretion of vascular endothelial growth factor (VEGF) was additionally quantified. RESULTS: Our hypothesis was validated by reduced expression of TJ proteins, occludin and claudin-5 together with significant barrier breakdown in iBCECs after only 24 h of co-culture, demonstrated by reduction in TEER from 1313 ± 265 Ω*cm2 to 712 ± 299 Ω*cm2 (iBCECs + U87-MG) and 762 ± 316 Ω*cm2 (iBCECs + U373-MG). Furthermore, 3D spheroids show more resistance to standard GBM chemotherapeutics in-vitro compared to 2D cultures. CONCLUSIONS: We demonstrate the establishment of a simplified, robust in-vitro BTB test system, with potential application in preclinical therapeutic screening and in studying GBM-induced pathological changes at the BBB.

5.
Biomedicines ; 12(7)2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39062123

RESUMEN

Human induced pluripotent stem cells (hiPSCs) represent a powerful tool to investigate neuropathological disorders in which the cells of interest are inaccessible, such as in the Charcot-Marie-Tooth disease (CMT), the most common inherited peripheral neuropathy. Developing appropriate cellular models becomes crucial in order to both study the disease's pathophysiology and test new therapeutic approaches. The generation of hiPS cellular models for disorders caused by a single nucleotide variation has been significantly improved following the development of CRISPR-based editing tools. In this study, we efficiently and quickly generated, by CRISPR editing, the two first hiPSCs cellular models carrying alterations involved in CMT4C, also called AR-CMTde-SH3TC2. This subtype of CMT is associated with alterations in the SH3TC2 gene and represents the most prevalent form of autosomal recessive demyelinating CMT. We aimed to develop models for two different SH3TC2 nonsense variants, c.211C>T, p.Gln71* and the most common AR-CMTde-SH3TC2 alteration, c.2860C>T, p.Arg954*. First, in order to determine the best CRISPR strategy to adopt on hiPSCs, we first tested a variety of sgRNAs combined with a selection of recent base editors using the conveniently cultivable and transfectable HEK-293T cell line. The chosen CRISPR base-editing strategy was then applied to hiPSCs derived from healthy individuals to generate isogenic CMT disease models with up to 93% editing efficiency. For point mutation generation, we first recommend to test your strategies on alternative cell line such as HEK-293T before hiPSCs to evaluate a variety of sgRNA-BE combinations, thus boosting the chance of achieving edited cellular clones with the hard-to-culture and to transfect hiPSCs.

6.
eNeuro ; 11(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39009447

RESUMEN

Generation of human induced pluripotent stem cell (hiPSC)-derived motor neurons (MNs) offers an unprecedented approach to modeling movement disorders such as dystonia and amyotrophic lateral sclerosis. However, achieving survival poses a significant challenge when culturing induced MNs, especially when aiming to reach late maturation stages. Utilizing hiPSC-derived motor neurons and primary mouse astrocytes, we assembled two types of coculture systems: direct coculturing of neurons with astrocytes and indirect coculture using culture inserts that physically separate neurons and astrocytes. Both systems significantly enhance neuron survival. Compared with these two systems, no significant differences in neurodevelopment, maturation, and survival within 3 weeks, allowing to prepare neurons at maturation stages. Using the indirect coculture system, we obtained highly pure MNs at the late mature stage from hiPSCs. Transcriptomic studies of hiPSC-derived MNs showed a typical neurodevelopmental switch in gene expression from the early immature stage to late maturation stages. Mature genes associated with neurodevelopment and synaptogenesis are highly enriched in MNs at late stages, demonstrating that these neurons achieve maturation. This study introduces a novel tool for the preparation of highly pure hiPSC-derived neurons, enabling the determination of neurological disease pathogenesis in neurons at late disease onset stages through biochemical approaches, which typically necessitate highly pure neurons. This advancement is particularly significant in modeling age-related neurodegeneration.


Asunto(s)
Astrocitos , Técnicas de Cocultivo , Células Madre Pluripotentes Inducidas , Neuronas Motoras , Células Madre Pluripotentes Inducidas/fisiología , Animales , Neuronas Motoras/fisiología , Ratones , Astrocitos/fisiología , Humanos , Diferenciación Celular/fisiología , Células Cultivadas , Neurogénesis/fisiología
7.
Cardiovasc Res ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38916487

RESUMEN

AIMS: Endothelial cell (EC) dysfunction plays a key role in the initiation and progression of cardiovascular disease. However, studying these disorders in ECs from patients is challenging, hence the use of human induced pluripotent stem cells (hiPSCs) and their in vitro differentiation into ECs represents a very promising approach. Still, the generation of hiPSC-derived ECs (hECs) remains demanding as a cocktail of growth factors and an intermediate purification step are required for hEC enrichment. Therefore, we probed the utility of a forward programming approach using transgenic hiPSC lines. METHODS AND RESULTS: We have used the transgenic hiPSC line PGP1 ETV2 iso2 to explore the in vitro differentiation of hECs via doxycycline-dependent induction of the transcription factor ETV2 and compared these with a standard differentiation protocol for hECs using non-transgenic control hiPSCs. The transgenic hECs were highly enriched without an intermediate purification step and expressed - as non-transgenic hECs and HUVECs - characteristic EC markers. The viability and yield of transgenic hECs were strongly improved by applying EC growth medium during differentiation. This protocol was successfully applied in two more transgenic hiPSC lines yielding reproducible results with low line-to-line variability. Transgenic hECs displayed typical functional properties, such as tube formation and LDL uptake, and a more mature phenotype than non-transgenic hECs. Transgenic hiPSCs preferentially differentiated into the arterial lineage, this was further enhanced by adding a high VEGF concentration to the medium. We also demonstrate that complexing lentivirus with magnetic nanoparticles and application of a magnetic field enables efficient transduction of transgenic hECs. CONCLUSIONS: We have established a highly efficient, cost-effective, and reproducible differentiation protocol for the generation of functional hECs via forward programming. The transgenic hECs can be genetically modified and are a powerful tool for disease modelling, tissue engineering, and translational purposes.

8.
Folia Neuropathol ; 62(1): 32-46, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38741435

RESUMEN

Human induced pluripotent stem cells (hiPSCs) are a potential source of somatic cells for cell therapies due to their ability to self-renew and differentiate into various cells of the body. To date, the clinical application of hiPSCs has been limited due to safety issues. The present study aims to standardize the safety procedure of the derivation of GMP-compliant induced pluripotent stem cell (iPSC) lines from human fibroblasts. The hiPSC lines were generated using the nonintegrative Sendai virus method to incorporate Yamanaka reprogramming factors (OCT3/4, SOX2, KLF4 and c-MYC) into cells. A constant temperature was maintained during the cell culture, including all stages of the culture after transduction with Sendai virus. Pluripotency was proved in six independently generated hiPSC lines from adult female (47 years old) and male (57 years old) donors' derived fibroblasts via alkaline phosphatase live (ALP) staining, qPCR, and immunocytochemistry. The hiPSC lines showed a gradual decrease in the presence of the virus with each subsequent passage, and this reduction was specific to the hiPSC line. The frequency and probability of chromosomal aberrations in hiPSCs were dependent on both the iPSC clone identity and sex of the donor. In summary, the generation of hiPSC for clinical applications requires safety standards application (biosafety protocol, quality control of hiPSC lines, viral and genetic integrity screening) from the first stages of the clonal selection of hiPSC from the same donor.


Asunto(s)
Células Madre Pluripotentes Inducidas , Factor 4 Similar a Kruppel , Virus Sendai , Humanos , Femenino , Masculino , Persona de Mediana Edad , Línea Celular , Fibroblastos , Diferenciación Celular/fisiología , Transducción Genética/métodos , Factores Sexuales
9.
ACS Appl Mater Interfaces ; 16(22): 28263-28275, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38788694

RESUMEN

Intervertebral disc degeneration (IDD) is a progressive condition and stands as one of the primary causes of low back pain. Cell therapy that uses nucleus pulposus (NP)-like cells derived from human induced pluripotent stem cells (hiPSCs) holds great promise as a treatment for IDD. However, the conventional two-dimensional (2D) monolayer cultures oversimplify cell-cell interactions, leading to suboptimal differentiation efficiency and potential loss of phenotype. While three-dimensional (3D) culture systems like Matrigel improve hiPSC differentiation efficiency, they are limited by animal-derived materials for translation, poorly defined composition, short-term degradation, and high cost. In this study, we introduce a new 3D scaffold fabricated using medical-grade chitosan with a high degree of deacetylation. The scaffold features a highly interconnected porous structure, near-neutral surface charge, and exceptional degradation stability, benefiting iPSC adhesion and proliferation. This scaffold remarkably enhances the differentiation efficiency and allows uninterrupted differentiation for up to 25 days without subculturing. Notably, cells differentiated on the chitosan scaffold exhibited increased cell survival rates and upregulated gene expression associated with extracellular matrix secretion under a chemically defined condition mimicking the challenging microenvironment of intervertebral discs. These characteristics qualify the chitosan scaffold-cell construct for direct implantation, serving as both a structural support and a cellular source for enhanced stem cell therapy for IDD.


Asunto(s)
Diferenciación Celular , Quitosano , Células Madre Pluripotentes Inducidas , Núcleo Pulposo , Andamios del Tejido , Quitosano/química , Diferenciación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Núcleo Pulposo/citología , Humanos , Andamios del Tejido/química , Degeneración del Disco Intervertebral/terapia , Degeneración del Disco Intervertebral/patología , Células Cultivadas , Supervivencia Celular/efectos de los fármacos
10.
Hum Cell ; 37(4): 1205-1214, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38762696

RESUMEN

Hypertrophic cardiomyopathy (HCM) is frequently caused by mutations in the MYPBC3 gene, which encodes the cardiac myosin-binding protein C (cMyBP-C). Most pathogenic variants in MYPBC3 are either nonsense mutations or result in frameshifts, suggesting that the primary disease mechanism involves reduced functional cMyBP-C protein levels within sarcomeres. However, a subset of MYPBC3 variants are missense mutations, and the molecular mechanisms underlying their pathogenicity remain elusive. Upon in vitro differentiation into cardiomyocytes, induced pluripotent stem cells (iPSCs) derived from HCM patients represent a valuable resource for disease modeling. In this study, we generated two iPSC lines from peripheral blood mononuclear cells (PBMCs) of a female with early onset and severe HCM linked to the MYBPC3: c.772G > A variant. Although this variant was initially classified as a missense mutation, recent studies indicate that it interferes with splicing and results in a frameshift. The generated iPSC lines exhibit a normal karyotype and display hallmark characteristics of pluripotency, including the ability to undergo trilineage differentiation. These novel iPSCs expand the existing repertoire of MYPBC3-mutated cell lines, broadening the spectrum of resources for exploring how diverse mutations induce HCM. They additionally offer a platform to study potential secondary genetic elements contributing to the pronounced disease severity observed in this individual.


Asunto(s)
Cardiomiopatía Hipertrófica , Proteínas Portadoras , Diferenciación Celular , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/etiología , Femenino , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Mutación Missense/genética , Índice de Severidad de la Enfermedad , Mutación/genética , Línea Celular , Mutación del Sistema de Lectura/genética , Leucocitos Mononucleares/metabolismo , Células Cultivadas
11.
Stem Cell Res Ther ; 15(1): 152, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38816767

RESUMEN

BACKGROUND: X-linked juvenile retinoschisis (XLRS) is an inherited disease caused by RS1 gene mutation, which leads to retinal splitting and visual impairment. The mechanism of RS1-associated retinal degeneration is not fully understood. Besides, animal models of XLRS have limitations in the study of XLRS. Here, we used human induced pluripotent stem cell (hiPSC)-derived retinal organoids (ROs) to investigate the disease mechanisms and potential treatments for XLRS. METHODS: hiPSCs reprogrammed from peripheral blood mononuclear cells of two RS1 mutant (E72K) XLRS patients were differentiated into ROs. Subsequently, we explored whether RS1 mutation could affect RO development and explore the effectiveness of RS1 gene augmentation therapy. RESULTS: ROs derived from RS1 (E72K) mutation hiPSCs exhibited a developmental delay in the photoreceptor, retinoschisin (RS1) deficiency, and altered spontaneous activity compared with control ROs. Furthermore, the delays in development were associated with decreased expression of rod-specific precursor markers (NRL) and photoreceptor-specific markers (RCVRN). Adeno-associated virus (AAV)-mediated gene augmentation with RS1 at the photoreceptor immature stage rescued the rod photoreceptor developmental delay in ROs with the RS1 (E72K) mutation. CONCLUSIONS: The RS1 (E72K) mutation results in the photoreceptor development delay in ROs and can be partially rescued by the RS1 gene augmentation therapy.


Asunto(s)
Proteínas del Ojo , Terapia Genética , Células Madre Pluripotentes Inducidas , Mutación , Organoides , Retina , Retinosquisis , Retinosquisis/genética , Retinosquisis/terapia , Retinosquisis/patología , Retinosquisis/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Terapia Genética/métodos , Organoides/metabolismo , Retina/metabolismo , Retina/patología , Masculino , Diferenciación Celular
12.
Methods Mol Biol ; 2803: 13-33, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38676882

RESUMEN

The adept and systematic differentiation of embryonic stem cells (ESCs) and human-induced pluripotent stem cells (hiPSCs) to diverse lineage-prone cell types involves crucial step-by-step process that mimics the vital strategic commitment phase that is usually observed during the process of embryo development. The development of precise tissue-specific cell types from these stem cells indeed plays an important role in the advancement of imminent stem cell-based therapeutic strategies. Therefore, the usage of hiPSC-derived cell types for subsequent cardiovascular disease modeling, drug screening, and therapeutic drug development undeniably entails an in-depth understanding of each and every step to proficiently stimulate these stem cells into desired cardiomyogenic lineage. Thus, to accomplish this definitive and decisive fate, it is essential to efficiently induce the mesoderm or pre-cardiac mesoderm, succeeded by the division of cells into cardiovascular and ultimately ensuing with the cardiomyogenic lineage outcome. This usually commences from the earliest phases of pluripotent cell induction. In this chapter, we discuss our robust and reproducible step-wise protocol that will describe the subtype controlled, precise lineage targeted standardization of activin/nodal, and BMP signaling molecules/cytokines, for the efficient differentiation of ventricular cardiomyocytes from hiPSCs via the embryoid body method. In addition, we also describe techniques to dissociate hiPSCs, hiPSC-derived early cardiomyocytes for mesoderm and pre-cardiac mesoderm assessment, and hiPSC-derived cardiomyocytes for early and mature markers assessment.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Humanos , Activinas/farmacología , Activinas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Técnicas de Cultivo de Célula/métodos , Linaje de la Célula , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteína Nodal/metabolismo , Transducción de Señal
13.
ACS Biomater Sci Eng ; 10(5): 3218-3231, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38593429

RESUMEN

Spinal cord organoids are of significant value in the research of spinal cord-related diseases by simulating disease states, thereby facilitating the development of novel therapies. However, the complexity of spinal cord structure and physiological functions, along with the lack of human-derived inducing components, presents challenges in the in vitro construction of human spinal cord organoids. Here, we introduce a novel human decellularized placenta-derived extracellular matrix hydrogel (DPECMH) and, combined with a new induction protocol, successfully construct human spinal cord organoids. The human placenta-sourced decellularized extracellular matrix (dECM), verified through hematoxylin and eosin staining, DNA quantification, and immunofluorescence staining, retained essential ECM components such as elastin, fibronectin, type I collagen, laminin, and so forth. The temperature-sensitive hydrogel made from human placenta dECM demonstrated good biocompatibility and promoted the differentiation of human induced pluripotent stem cell (hiPSCs)-derived spinal cord organoids into neurons. It displayed enhanced expression of laminar markers in comparison to Matrigel and showed higher expression of laminar markers compared to Matrigel, accelerating the maturation process of spinal cord organoids and demonstrating its potential as an organoid culture substrate. DPECMH has the potential to replace Matrigel as the standard additive for human spinal cord organoids, thus advancing the development of spinal cord organoid culture protocols and their application in the in vitro modeling of spinal cord-related diseases.


Asunto(s)
Diferenciación Celular , Matriz Extracelular Descelularizada , Hidrogeles , Células Madre Pluripotentes Inducidas , Organoides , Placenta , Médula Espinal , Humanos , Organoides/citología , Organoides/metabolismo , Organoides/efectos de los fármacos , Femenino , Placenta/citología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Embarazo , Hidrogeles/química , Hidrogeles/farmacología , Médula Espinal/citología , Médula Espinal/metabolismo , Diferenciación Celular/efectos de los fármacos , Matriz Extracelular Descelularizada/farmacología , Matriz Extracelular Descelularizada/química , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Laminina/farmacología , Laminina/química
14.
Methods Mol Biol ; 2803: 35-48, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38676883

RESUMEN

The lack of a precise noninvasive, clinical evaluation method for cardiac fibrosis hinders the development of successful treatments that can effectively work in physiological settings, where tissues and organs are interconnected and moderating drug responses. To address this challenge and advance personalized medicine, researchers have turned to human-induced pluripotent stem (iPS) cells, which can be differentiated to resemble the human heart in terms of structure, function and cellular composition. In this chapter, we present an assay protocol that uses these iPS cells to generate heart organoids for the in vitro evaluation of cardiac fibrosis. By establishing this biological platform, we pave the way for conducting phenotype evaluation and treatment screening in a multiscale approach, aiming to discover effective interventions for the treatment of cardiac fibrosis.


Asunto(s)
Diferenciación Celular , Fibrosis , Células Madre Pluripotentes Inducidas , Organoides , Humanos , Células Madre Pluripotentes Inducidas/citología , Organoides/patología , Organoides/citología , Miocardio/patología , Miocardio/citología , Técnicas de Cultivo de Célula/métodos , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología , Células Cultivadas
15.
J Neurovirol ; 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38600307

RESUMEN

The human immunodeficiency virus (HIV) epidemic is an ongoing global health problem affecting 38 million people worldwide with nearly 1.6 million new infections every year. Despite the advent of combined antiretroviral therapy (cART), a large percentage of people with HIV (PWH) still develop neurological deficits, grouped into the term of HIV-associated neurocognitive disorders (HAND). Investigating the neuropathology of HIV is important for understanding mechanisms associated with cognitive impairment seen in PWH. The major obstacle for studying neuroHIV is the lack of suitable in vitro human culture models that could shed light into the HIV-CNS interactions. Recent advances in induced pluripotent stem cell (iPSC) culture and 3D brain organoid systems have allowed the generation of 2D and 3D culture methods that possess a potential to serve as a model of neurotropic viral diseases, including HIV. In this study, we first generated and characterized several hiPSC lines from healthy human donor skin fibroblast cells. hiPSCs were then used for the generation of microglia-containing human cerebral organoids (hCOs). Once fully characterized, hCOs were infected with HIV-1 in the presence and absence of cART regimens and viral infection was studied by cellular, molecular/biochemical, and virological assays. Our results revealed that hCOs were productively infected with HIV-1 as evident by viral p24-ELISA in culture media, RT-qPCR and RNAscope analysis of viral RNA, as well as ddPCR analysis of proviral HIV-1 in genomic DNA samples. More interestingly, replication and gene expression of HIV-1 were also greatly suppressed by cART in hCOs as early as 7 days post-infections. Our results suggest that hCOs derived from hiPSCs support HIV-1 replication and gene expression and may serve as a unique platform to better understand neuropathology of HIV infection in the brain.

16.
Neuroscience ; 546: 1-19, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38522661

RESUMEN

Midbrain dopaminergic (mDA) neurons are significantly impaired in patients inflicted with Parkinson's disease (PD), subsequently affecting a variety of motor functions. There are four pathways through which dopamine elicits its function, namely, nigrostriatal, mesolimbic, mesocortical and tuberoinfundibular dopamine pathways. SHH and Wnt signalling pathways in association with favourable expression of a variety of genes, promotes the development and differentiation of mDA neurons in the brain. However, there is a knowledge gap regarding the complex signalling pathways involved in development of mDA neurons. hiPSC models have been acclaimed to be effective in generating complex disease phenotypes. These models mimic the microenvironment found in vivo thus ensuring maximum reliability. Further, a variety of therapeutic compounds can be screened using hiPSCs since they can be used to generate neurons that could carry an array of mutations associated with both familial and sporadic PD. Thus, culturing hiPSCs to study gene expression and dysregulation of cellular processes associated with PD can be useful in developing targeted therapies that will be a step towards halting disease progression.


Asunto(s)
Neuronas Dopaminérgicas , Células Madre Pluripotentes Inducidas , Mesencéfalo , Enfermedad de Parkinson , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Mesencéfalo/metabolismo , Mesencéfalo/patología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Diferenciación Celular/fisiología
17.
J Neurovirol ; 30(2): 131-145, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38478163

RESUMEN

The neurogenic niches within the central nervous system serve as essential reservoirs for neural precursor cells (NPCs), playing a crucial role in neurogenesis. However, these NPCs are particularly vulnerable to infection by the herpes simplex virus 1 (HSV-1). In the present study, we investigated the changes in the transcriptome of NPCs in response to HSV-1 infection using bulk RNA-Seq, compared to those of uninfected samples, at different time points post infection and in the presence or absence of antivirals. The results showed that NPCs upon HSV-1 infection undergo a significant dysregulation of genes playing a crucial role in aspects of neurogenesis, including genes affecting NPC proliferation, migration, and differentiation. Our analysis revealed that the CREB signaling, which plays a crucial role in the regulation of neurogenesis and memory consolidation, was the most consistantly downregulated pathway, even in the presence of antivirals. Additionally, cholesterol biosynthesis was significantly downregulated in HSV-1-infected NPCs. The findings from this study, for the first time, offer insights into the intricate molecular mechanisms that underlie the neurogenesis impairment associated with HSV-1 infection.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Células-Madre Neurales , Neurogénesis , RNA-Seq , Transcriptoma , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Células-Madre Neurales/virología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Animales , Herpes Simple/genética , Herpes Simple/virología , Herpes Simple/metabolismo , Antivirales/farmacología , Diferenciación Celular , Ratones , Transducción de Señal , Colesterol/metabolismo , Proliferación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Regulación de la Expresión Génica , Movimiento Celular
18.
J Neurosci ; 44(15)2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38438257

RESUMEN

DYT1 dystonia is a debilitating neurological movement disorder, and it represents the most frequent and severe form of hereditary primary dystonia. There is currently no cure for this disease due to its unclear pathogenesis. In our previous study utilizing patient-specific motor neurons (MNs), we identified distinct cellular deficits associated with the disease, including a deformed nucleus, disrupted neurodevelopment, and compromised nucleocytoplasmic transport (NCT) functions. However, the precise molecular mechanisms underlying these cellular impairments have remained elusive. In this study, we revealed the genome-wide changes in gene expression in DYT1 MNs through transcriptomic analysis. We found that those dysregulated genes are intricately involved in neurodevelopment and various biological processes. Interestingly, we identified that the expression level of RANBP17, a RAN-binding protein crucial for NCT regulation, exhibited a significant reduction in DYT1 MNs. By manipulating RANBP17 expression, we further demonstrated that RANBP17 plays an important role in facilitating the nuclear transport of both protein and transcript cargos in induced human neurons. Excitingly, the overexpression of RANBP17 emerged as a substantial mitigating factor, effectively restoring impaired NCT activity and rescuing neurodevelopmental deficits observed in DYT1 MNs. These findings shed light on the intricate molecular underpinnings of impaired NCT in DYT1 neurons and provide novel insights into the pathophysiology of DYT1 dystonia, potentially leading to the development of innovative treatment strategies.


Asunto(s)
Distonía Muscular Deformante , Distonía , Trastornos Distónicos , Proteína de Unión al GTP ran , Humanos , Transporte Activo de Núcleo Celular , Chaperonas Moleculares/genética , Neuronas Motoras/metabolismo
19.
ACS Chem Neurosci ; 15(6): 1265-1275, 2024 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-38421952

RESUMEN

Alzheimer's disease (AD) is associated with the aggregation of amyloid ß (Aß) and tau proteins. Why ApoE variants are significant genetic risk factors remains a major unsolved puzzle in understanding AD, although intracellular interactions with ApoE are suspected to play a role. Here, we show that specific changes in the fluorescence lifetime of fluorescently tagged small Aß oligomers in rat brain cells correlate with the cellular ApoE content. An inhibitor of the Aß-ApoE interaction suppresses these changes and concomitantly reduces Aß toxicity in a dose-dependent manner. Single-molecule techniques show changes both in the conformation and in the stoichiometry of the oligomers. Neural stem cells derived from hiPSCs of Alzheimer's patients also exhibit these fluorescence lifetime changes. We infer that intracellular interaction with ApoE modifies the N-terminus of the Aß oligomers, inducing changes in their stoichiometry, membrane affinity, and toxicity. These changes can be directly imaged in live cells and can potentially be used as a rapid and quantitative cellular assay for AD drug discovery.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Humanos , Ratas , Animales , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas tau/metabolismo
20.
Int J Biol Sci ; 20(4): 1160-1179, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38385067

RESUMEN

There is an urgent need for novel therapies to treat end-stage liver disease due to the shortage of available organs. Although cell transplantation holds considerable promise, its availability is limited due to the low engrafted cell mass and lack of unifying cell transplantation strategies. Here, we optimally established human induced pluripotent stem cell-derived functional hepatobiliary organoids (HBOs) based on our previous research and transplanted them into a monkey model via liver subcapsular and submesenteric transplantation routes to assess their potential clinical application. Our study revealed that HBO transplantation could safely and effectively improve hepatoprotection effects by antiapoptotic and antifibrotic agents. In addition, we also discovered that while multiple HBO transplantation pathways may have a shared effector mechanism, their respective treatment approaches have distinct advantages. Transplantation of HBOs could promote the high expression of CTSV in hepatic sinusoid endothelial cells, which might halt the progression of hepatic sinusoidal capillarization and liver fibrosis. Liver subcapsular transplants had stronger pro-CTSV upregulation than HBO submesenteric transplants, which could be attributed to naturally high CTSV expression in HBOs. Interestingly, both transplantation routes of HBOs were targeted the injured liver and triggered a new pattern of ductular reaction to alleviate the degree of liver fibrosis by surrounding the area with CK19-positive labeled cells. These residing, homing and repairing effects might be related to the high expression of MMP family genes. By promoting a unique pattern of ductular reactions, submesenteric HBO transplantation has a more representative antifibrotic impact than liver subcapsular transplantation. Altogether, our data strongly imply that the treatment of severe liver diseases with liver subcapsular and submesenteric transplantation of HBOs may be clinically effective and safe. These findings provide new insight into HBOs for further experimental and clinical validation.


Asunto(s)
Colestasis , Células Madre Pluripotentes Inducidas , Animales , Humanos , Células Endoteliales , Cirrosis Hepática/inducido químicamente , Hígado/patología , Colestasis/patología , Organoides , Primates
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA