Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Small ; 18(36): e2202309, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35948487

RESUMEN

Functional microgels are preferred stem cell carriers due to the ease of delivery through minimally invasive injection and seamless integration with the surrounding host tissue. A biostimulatory nanofiber-hydrogel composite (NHC) has been previously developed through covalently crosslinking a hyaluronic acid hydrogel network with surface-functionalized poly (ε-caprolactone) nanofiber fragments. The NHC mimics the microarchitecture of native soft tissue matrix, showing enhanced cell infiltration, immunomodulation, and proangiogenic properties. Here, injectability of the pre-formed NHC is improved by mechanical fragmentation, making it into micro-fragmented NHC (mfNHC) in a granular gel form as a stem cell carrier to deliver mesenchymal stem cells (MSCs) for soft tissue remodeling. The mfNHC shows a similar storage modulus but a significantly reduced injection force, as compared with the corresponding bulk NHC. When injected subcutaneously in a rat model, mfNHC-MSC constructs initiate an elevated level of host macrophage infiltration, more pro-regenerative polarization, and subsequently, improved angiogenesis and adipogenesis response when compared to mfNHC alone. A similar trend of host cell infiltration and pro-angiogenic response is detected in a swine model with a larger volume injection. These results suggest a strong potential for use of the mfNHC as an injectable carrier for cell delivery and soft tissue remodeling.


Asunto(s)
Células Madre Mesenquimatosas , Nanofibras , Animales , Ácido Hialurónico , Hidrogeles , Inyecciones , Células Madre Mesenquimatosas/fisiología , Ratas , Porcinos , Ingeniería de Tejidos/métodos
2.
Proc Natl Acad Sci U S A ; 111(28): 10209-14, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24987120

RESUMEN

Excessive poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) activation kills cells via a cell-death process designated "parthanatos" in which PAR induces the mitochondrial release and nuclear translocation of apoptosis-inducing factor to initiate chromatinolysis and cell death. Accompanying the formation of PAR are the reduction of cellular NAD(+) and energetic collapse, which have been thought to be caused by the consumption of cellular NAD(+) by PARP-1. Here we show that the bioenergetic collapse following PARP-1 activation is not dependent on NAD(+) depletion. Instead PARP-1 activation initiates glycolytic defects via PAR-dependent inhibition of hexokinase, which precedes the NAD(+) depletion in N-methyl-N-nitroso-N-nitroguanidine (MNNG)-treated cortical neurons. Mitochondrial defects are observed shortly after PARP-1 activation and are mediated largely through defective glycolysis, because supplementation of the mitochondrial substrates pyruvate and glutamine reverse the PARP-1-mediated mitochondrial dysfunction. Depleting neurons of NAD(+) with FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, does not alter glycolysis or mitochondrial function. Hexokinase, the first regulatory enzyme to initiate glycolysis by converting glucose to glucose-6-phosphate, contains a strong PAR-binding motif. PAR binds to hexokinase and inhibits hexokinase activity in MNNG-treated cortical neurons. Preventing PAR formation with PAR glycohydrolase prevents the PAR-dependent inhibition of hexokinase. These results indicate that bioenergetic collapse induced by overactivation of PARP-1 is caused by PAR-dependent inhibition of glycolysis through inhibition of hexokinase.


Asunto(s)
Corteza Cerebral/enzimología , Glucólisis/fisiología , Mitocondrias/enzimología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Acrilamidas/farmacología , Animales , Células Cultivadas , Corteza Cerebral/citología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Glucosa/metabolismo , Glucosa-6-Fosfato/metabolismo , Glucólisis/efectos de los fármacos , Hexoquinasa/metabolismo , Metilnitronitrosoguanidina/farmacología , Ratones , NAD/metabolismo , Neuronas/citología , Piperidinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1
3.
Am J Physiol Endocrinol Metab ; 306(2): E150-6, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24253050

RESUMEN

A splice form of IGF-1, IGF-1Eb, is upregulated after exercise or injury. Physiological responses have been ascribed to the 24-amino acid COOH-terminal peptide that is cleaved from the NH3-terminal 70-amino acid mature IGF-1 protein. This COOH-terminal peptide was termed "mechano-growth factor" (MGF). Activities claimed for the MGF peptide included enhancing muscle satellite cell proliferation and delaying myoblast fusion. As such, MGF could represent a promising strategy to improve muscle regeneration. Thus, at our two pharmaceutical companies, we attempted to reproduce the claimed effect of MGF peptides on human and mouse muscle myoblast proliferation and differentiation in vitro. Concentrations of peptide up to 500 ng/ml failed to increase the proliferation of C2C12 cells or primary human skeletal muscle myoblasts. In contrast, all cell types exhibited a proliferative response to mature IGF-1 or full-length IGF-1Eb. MGF also failed to inhibit the differentiation of myoblasts into myotubes. To address whether the response to MGF was lost in these tissue culture lines, we measured proliferation and differentiation of primary mouse skeletal muscle stem cells exposed to MGF. This, too, failed to demonstrate a significant effect. Finally, we tested whether MGF could alter a separate documented in vitro effect of the peptide, activation of p-ERK, but not p-Akt, in cardiac myocytes. Although a robust response to IGF-1 was observed, there were no demonstrated activating responses from the native or a stabilized MGF peptide. These results call in to question whether there is a physiological role for MGF.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Mioblastos/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Factor I del Crecimiento Similar a la Insulina/química , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones , Mioblastos/fisiología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Cultivo Primario de Células , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Células Madre/fisiología
4.
J Immunol Methods ; 519: 113514, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37353001

RESUMEN

Peripheral blood mononuclear cells (PBMCs) are commonly isolated from whole blood samples in clinical trials. Isolated PBMCs can be cryopreserved for use in downstream assays such as flow cytometry, single-cell RNA sequencing (scRNA-seq) and enzyme-linked immunosorbent spot (ELISpot) assays to aid understanding of disease biology and treatment effects, and biomarker identification. However, due to logistical practicalities, delays from blood collection to PBMC processing may exceed 24 h, which can potentially affect PBMC function and, ultimately, downstream assay results. Whole blood samples from 20 healthy adults were collected and incubated at 20-25 °C for 2-48 h before PBMC processing. PBMC viability was measured, and flow cytometry immunophenotyping, scRNA-seq and ELISpot were performed following increasing PBMC processing delays. The RosetteSep™ granulocyte depletion kit was used to evaluate the impact of granulocyte contamination following processing delay. Processed scRNA-seq reads were used to identify cell clusters based on marker genes. scRNA-seq data was further used to determine gene expression correlation and pathway activity score in major PBMC cell types (T cells, B cells, natural killer cells, monocytes and dendritic cells) between PBMC preparations subjected to shorter (2-4 h) and longer (8-48 h) processing delays. ELISpot assays evaluated the impact of processing delays on the number of interferon-γ (IFN-γ) secreting cells from ex vivo stimulated PBMCs. PBMC viability was reduced after a 48-h processing delay. Flow cytometry showed that granulocyte contamination of PBMCs increased after 24 h. Cluster analysis of scRNA-seq data identified 23 immune cell type gene expression clusters that were not significantly changed upon granulocyte depletion. Gene expression correlations across the major PBMC cell types were < 0.8 after 24 h of delay compared with 2 or 4 h of delay. Inflammatory, proliferation and signaling pathway activities increased, whereas IFN-γ and metabolic pathway activities decreased with increasing PBMC processing delays. The number of IFN-γ secreting cells trended towards a reduction as PBMC processing delays increased. PBMC processing delays should be minimised when designing clinical trials to reduce outcome variability in downstream assays. Ideally clinical trial sites should have on-site PBMC processing capabilities or be located close to such facilities.


Asunto(s)
Leucocitos Mononucleares , Linfocitos T , Adulto , Humanos , Interferón gamma/farmacología , Células Asesinas Naturales , Genómica
5.
iScience ; 26(3): 106156, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36852281

RESUMEN

Promoting myelination capacity of endogenous oligodendrocyte precursor cells (OPCs) is a promising therapeutic approach for CNS demyelinating disorders such as Multiple Sclerosis (MS). To aid in the discovery of myelination-promoting compounds, we generated a genome-engineered human pluripotent stem cell (hPSC) line that consists of three reporters: identification-and-purification tag, GFP, and secreted-NanoLuc, driven by the endogenous PDGFRA, PLP1, and MBP genes, respectively. Using this cell line, we established a high-throughput drug screening platform and performed a small-molecule screen, which identified at least two myelination-promoting small-molecule (Ro1138452 and SR2211) that target prostacyclin (IP) receptor and retinoic acid receptor-related orphan receptor γ (RORγ), respectively. Single-cell-transcriptomic analysis of differentiating OPCs treated with these molecules further confirmed that they promote oligodendrocyte differentiation and revealed several pathways that are potentially modulated by them. The molecules and their target pathways provide promising targets for the possible development of remyelination-based therapy for MS and other demyelinating disorders.

6.
Adv Healthc Mater ; 10(7): e2001689, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33433956

RESUMEN

Mesenchymal stem cells (MSCs) have been widely studied as a versatile cell source for tissue regeneration and remodeling due to their potent bioactivity, which includes modulation of inflammation response, macrophage polarization toward proregenerative lineage, promotion of angiogenesis, and reduction in fibrosis. This review focuses on profiling the effects of paracrine signals of MSCs, commonly referred to as the secretome, and highlighting the various engineering approaches to tune the MSC secretome. Recent advances in biomaterials-based therapeutic strategies for delivery of MSCs and MSC-derived secretome in the form of extracellular vesicles are discussed, along with their advantages and challenges.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Fibrosis , Humanos , Comunicación Paracrina , Cicatrización de Heridas
7.
Mol Neurodegener ; 16(1): 25, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33853653

RESUMEN

BACKGROUND: Apoptosis-inducing factor (AIF), as a mitochondrial flavoprotein, plays a fundamental role in mitochondrial bioenergetics that is critical for cell survival and also mediates caspase-independent cell death once it is released from mitochondria and translocated to the nucleus under ischemic stroke or neurodegenerative diseases. Although alternative splicing regulation of AIF has been implicated, it remains unknown which AIF splicing isoform will be induced under pathological conditions and how it impacts mitochondrial functions and neurodegeneration in adult brain. METHODS: AIF splicing induction in brain was determined by multiple approaches including 5' RACE, Sanger sequencing, splicing-specific PCR assay and bottom-up proteomic analysis. The role of AIF splicing in mitochondria and neurodegeneration was determined by its biochemical properties, cell death analysis, morphological and functional alterations and animal behavior. Three animal models, including loss-of-function harlequin model, gain-of-function AIF3 knockin model and conditional inducible AIF splicing model established using either Cre-loxp recombination or CRISPR/Cas9 techniques, were applied to explore underlying mechanisms of AIF splicing-induced neurodegeneration. RESULTS: We identified a nature splicing AIF isoform lacking exons 2 and 3 named as AIF3. AIF3 was undetectable under physiological conditions but its expression was increased in mouse and human postmortem brain after stroke. AIF3 splicing in mouse brain caused enlarged ventricles and severe neurodegeneration in the forebrain regions. These AIF3 splicing mice died 2-4 months after birth. AIF3 splicing-triggered neurodegeneration involves both mitochondrial dysfunction and AIF3 nuclear translocation. We showed that AIF3 inhibited NADH oxidase activity, ATP production, oxygen consumption, and mitochondrial biogenesis. In addition, expression of AIF3 significantly increased chromatin condensation and nuclear shrinkage leading to neuronal cell death. However, loss-of-AIF alone in harlequin or gain-of-AIF3 alone in AIF3 knockin mice did not cause robust neurodegeneration as that observed in AIF3 splicing mice. CONCLUSIONS: We identified AIF3 as a disease-inducible isoform and established AIF3 splicing mouse model. The molecular mechanism underlying AIF3 splicing-induced neurodegeneration involves mitochondrial dysfunction and AIF3 nuclear translocation resulting from the synergistic effect of loss-of-AIF and gain-of-AIF3. Our study provides a valuable tool to understand the role of AIF3 splicing in brain and a potential therapeutic target to prevent/delay the progress of neurodegenerative diseases.


Asunto(s)
Empalme Alternativo , Factor Inductor de la Apoptosis/fisiología , Mitocondrias/metabolismo , Degeneración Nerviosa/genética , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Factor Inductor de la Apoptosis/deficiencia , Factor Inductor de la Apoptosis/genética , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Exones/genética , Femenino , Lóbulo Frontal/química , Mutación con Ganancia de Función , Edición Génica , Técnicas de Sustitución del Gen , Humanos , Lactante , Recién Nacido , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Persona de Mediana Edad , Neuronas/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología
8.
Nat Commun ; 12(1): 652, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33510160

RESUMEN

Injury and loss of oligodendrocytes can cause demyelinating diseases such as multiple sclerosis. To improve our understanding of human oligodendrocyte development, which could facilitate development of remyelination-based treatment strategies, here we describe time-course single-cell-transcriptomic analysis of developing human stem cell-derived oligodendrocyte-lineage-cells (hOLLCs). The study includes hOLLCs derived from both genome engineered embryonic stem cell (ESC) reporter cells containing an Identification-and-Purification tag driven by the endogenous PDGFRα promoter and from unmodified induced pluripotent (iPS) cells. Our analysis uncovers substantial transcriptional heterogeneity of PDGFRα-lineage hOLLCs. We discover sub-populations of human oligodendrocyte progenitor cells (hOPCs) including a potential cytokine-responsive hOPC subset, and identify candidate regulatory genes/networks that define the identity of these sub-populations. Pseudotime trajectory analysis defines developmental pathways of oligodendrocytes vs astrocytes from PDGFRα-expressing hOPCs and predicts differentially expressed genes between the two lineages. In addition, pathway enrichment analysis followed by pharmacological intervention of these pathways confirm that mTOR and cholesterol biosynthesis signaling pathways are involved in maturation of oligodendrocytes from hOPCs.


Asunto(s)
Heterogeneidad Genética , Variación Genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Colesterol/biosíntesis , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Precursoras de Oligodendrocitos/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
9.
Adv Mater ; 31(23): e1807359, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30968468

RESUMEN

T cell therapies require the removal and culture of T cells ex vivo to expand several thousand-fold. However, these cells often lose the phenotype and cytotoxic functionality for mediating effective therapeutic responses. The extracellular matrix (ECM) has been used to preserve and augment cell phenotype; however, it has not been applied to cellular immunotherapies. Here, a hyaluronic acid (HA)-based hydrogel is engineered to present the two stimulatory signals required for T-cell activation-termed an artificial T-cell stimulating matrix (aTM). It is found that biophysical properties of the aTM-stimulatory ligand density, stiffness, and ECM proteins-potentiate T cell signaling and skew phenotype of both murine and human T cells. Importantly, the combination of the ECM environment and mechanically sensitive TCR signaling from the aTM results in a rapid and robust expansion of rare, antigen-specific CD8+ T cells. Adoptive transfer of these tumor-specific cells significantly suppresses tumor growth and improves animal survival compared with T cells stimulated by traditional methods. Beyond immediate immunotherapeutic applications, demonstrating the environment influences the cellular therapeutic product delineates the importance of the ECM and provides a case study of how to engineer ECM-mimetic materials for therapeutic immune stimulation in the future.


Asunto(s)
Células Artificiales/citología , Ingeniería Celular/métodos , Inmunoterapia/métodos , Linfocitos T/citología , Traslado Adoptivo , Animales , Células Artificiales/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Matriz Extracelular/química , Humanos , Ácido Hialurónico/química , Hidrogeles , Ligandos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Science ; 354(6308)2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27846469

RESUMEN

Inhibition or genetic deletion of poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) is protective against toxic insults in many organ systems. The molecular mechanisms underlying PARP-1-dependent cell death involve release of mitochondrial apoptosis-inducing factor (AIF) and its translocation to the nucleus, which results in chromatinolysis. We identified macrophage migration inhibitory factor (MIF) as a PARP-1-dependent AIF-associated nuclease (PAAN). AIF was required for recruitment of MIF to the nucleus, where MIF cleaves genomic DNA into large fragments. Depletion of MIF, disruption of the AIF-MIF interaction, or mutation of glutamic acid at position 22 in the catalytic nuclease domain blocked MIF nuclease activity and inhibited chromatinolysis, cell death induced by glutamate excitotoxicity, and focal stroke. Inhibition of MIF's nuclease activity is a potential therapeutic target for diseases caused by excessive PARP-1 activation.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Apoptosis , División del ADN , Daño del ADN , ADN de Cadena Simple/metabolismo , Desoxirribonucleasas/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Factor Inductor de la Apoptosis/genética , Secuencia de Bases , Dominio Catalítico , Núcleo Celular/enzimología , Cromatina/metabolismo , Daño del ADN/genética , Fragmentación del ADN , Desoxirribonucleasas/química , Desoxirribonucleasas/genética , Ácido Glutámico/química , Ácido Glutámico/genética , Ácido Glutámico/toxicidad , Células HeLa , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Noqueados , Mitocondrias/enzimología , Mutación , Neuronas/enzimología , Conformación de Ácido Nucleico , Estrés Oxidativo , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/genética
11.
PLoS One ; 9(2): e90398, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24587351

RESUMEN

Satellite cells are the chief contributor to skeletal muscle growth and regeneration. The study of mouse satellite cells has accelerated in recent years due to technical advancements in the isolation of these cells. The study of human satellite cells has lagged and thus little is known about how the biology of mouse and human satellite cells compare. We developed a flow cytometry-based method to prospectively isolate human skeletal muscle progenitors from the satellite cell pool using positive and negative selection markers. Results show that this pool is enriched in PAX7 expressing cells that possess robust myogenic potential including the ability to give rise to de novo muscle in vivo. We compared mouse and human satellite cells in culture and identify differences in the elaboration of the myogenic genetic program and in the sensitivity of the cells to cytokine stimulation. These results indicate that not all mechanisms regulating mouse satellite cell activation are conserved in human satellite cells and that such differences may impact the clinical translation of therapeutics validated in mouse models. Thus, the findings of this study are relevant to developing therapies to combat muscle disease.


Asunto(s)
Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Adolescente , Adulto , Animales , Biomarcadores/metabolismo , Femenino , Citometría de Flujo , Expresión Génica , Humanos , Masculino , Ratones , Persona de Mediana Edad , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Proteína MioD/genética , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Células Satélite del Músculo Esquelético/citología , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA