Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 548(7667): 334-337, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28783732

RESUMEN

Cells in healthy tissues acquire mutations with surprising frequency. Many of these mutations are associated with abnormal cellular behaviours such as differentiation defects and hyperproliferation, yet fail to produce macroscopically detectable phenotypes. It is currently unclear how the tissue remains phenotypically normal, despite the presence of these mutant cells. Here we use intravital imaging to track the fate of mouse skin epithelium burdened with varying numbers of activated Wnt/ß-catenin stem cells. We show that all resulting growths that deform the skin tissue architecture regress, irrespective of their size. Wild-type cells are required for the active elimination of mutant cells from the tissue, while utilizing both endogenous and ectopic cellular behaviours to dismantle the aberrant structures. After regression, the remaining structures are either completely eliminated or converted into functional skin appendages in a niche-dependent manner. Furthermore, tissue aberrancies generated from oncogenic Hras, and even mutation-independent deformations to the tissue, can also be corrected, indicating that this tolerance phenomenon reflects a conserved principle in the skin. This study reveals an unanticipated plasticity of the adult skin epithelium when faced with mutational and non-mutational insult, and elucidates the dynamic cellular behaviours used for its return to a homeostatic state.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Homeostasis , Mutación , Fenotipo , Piel/citología , Animales , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células Madre/citología , Células Madre/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
2.
Nano Lett ; 14(10): 5677-86, 2014 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-25226349

RESUMEN

The rapidly approaching smart/wearable energy era necessitates advanced rechargeable power sources with reliable electrochemical properties and versatile form factors. Here, as a unique and promising energy storage system to address this issue, we demonstrate a new class of heterolayered, one-dimensional (1D) nanobuilding block mat (h-nanomat) battery based on unitized separator/electrode assembly (SEA) architecture. The unitized SEAs consist of wood cellulose nanofibril (CNF) separator membranes and metallic current collector-/polymeric binder-free electrodes comprising solely single-walled carbon nanotube (SWNT)-netted electrode active materials (LiFePO4 (cathode) and Li4Ti5O12 (anode) powders are chosen as model systems to explore the proof of concept for h-nanomat batteries). The nanoporous CNF separator plays a critical role in securing the tightly interlocked electrode-separator interface. The SWNTs in the SEAs exhibit multifunctional roles as electron conductive additives, binders, current collectors and also non-Faradaic active materials. This structural/physicochemical uniqueness of the SEAs allows significant improvements in the mass loading of electrode active materials, electron transport pathways, electrolyte accessibility and misalignment-proof of separator/electrode interface. As a result, the h-nanomat batteries, which are easily fabricated by stacking anode SEA and cathode SEA, provide unprecedented advances in the electrochemical performance, shape flexibility and safety tolerance far beyond those achievable with conventional battery technologies. We anticipate that the h-nanomat batteries will open 1D nanobuilding block-driven new architectural design/opportunity for development of next-generation energy storage systems.

3.
Front Physiol ; 15: 1397067, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38711955

RESUMEN

The hair follicle is a dynamic mini-organ that has specialized cycles and architectures with diverse cell types to form hairs. Previous studies for several decades have investigated morphogenesis and signaling pathways during embryonic development and adult hair cycles in both mouse and human skin. In particular, hair follicle stem cells and mesenchymal niches received major attention as key players, and their roles and interactions were heavily revealed. Although resident and circulating immune cells affect cellular function and interactions in the skin, research on immune cells has mainly received attention on diseases rather than development or homeostasis. Recently, many studies have suggested the functional roles of diverse immune cells as a niche for hair follicles. Here, we will review recent findings about immune niches for hair follicles and provide insight into mechanisms of hair growth and diseases.

4.
Int J Stem Cells ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38191522

RESUMEN

The multiple layers of the skin cover and protect our entire body. Among the skin layers, the epidermis is in direct contact with the outer environment and serves as the first line of defense. The epidermis functions as a physical and immunological barrier. To maintain barrier function, the epidermis continually regenerates and repairs itself when injured. Interactions between tissue-resident immune cells and epithelial cells are essential to sustain epidermal regeneration and repair. In this review, we will dissect the crosstalk between epithelial cells and specific immune cell populations located in the epidermis during homeostasis and wound repair. In addition, we will analyze the contribution of dysregulated immune-epithelial interactions in chronic inflammatory diseases.

5.
Stem Cell Reports ; 19(3): 317-330, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38335962

RESUMEN

Congenital heart defects are the most prevalent human birth defects, and their incidence is exacerbated by maternal health conditions, such as diabetes during the first trimester (pregestational diabetes). Our understanding of the pathology of these disorders is hindered by a lack of human models and the inaccessibility of embryonic tissue. Using an advanced human heart organoid system, we simulated embryonic heart development under pregestational diabetes-like conditions. These organoids developed pathophysiological features observed in mouse and human studies before, including ROS-mediated stress and cardiomyocyte hypertrophy. scRNA-seq revealed cardiac cell-type-specific dysfunction affecting epicardial and cardiomyocyte populations and alterations in the endoplasmic reticulum and very-long-chain fatty acid lipid metabolism. Imaging and lipidomics confirmed these findings and showed that dyslipidemia was linked to fatty acid desaturase 2 mRNA decay dependent on IRE1-RIDD signaling. Targeting IRE1 or restoring lipid levels partially reversed the effects of pregestational diabetes, offering potential preventive and therapeutic strategies in humans.


Asunto(s)
Cardiomiopatías , Diabetes Mellitus , Cardiopatías Congénitas , Humanos , Ratones , Animales , Cardiopatías Congénitas/patología , Estrés del Retículo Endoplásmico/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Organoides/metabolismo , Lípidos
6.
Stem Cells ; 30(5): 876-87, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22311737

RESUMEN

CD49f (integrin subunit α6) regulates signaling pathways in a variety of cellular activities. However, the role of CD49f in regulating the differentiation and pluripotency of stem cells has not been fully investigated. Therefore, in this study, human mesenchymal stem cells (hMSCs) were induced to form spheres under nonadherent culture conditions, and we found that the CD49f-positive population was enriched in MSC spheres compared with MSCs in a monolayer. The expression of CD49f regulated the ability of hMSCs to form spheres and was associated with an activation of the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway. Furthermore, the forced expression of CD49f modulated the proliferation and differentiation potentials of hMSCs through prolonged activation of PI3K/AKT and suppressed the level of p53. We showed that the pluripotency factors OCT4 and SOX2 were recruited to the putative promoter region of CD49f, indicating that OCT4 and SOX2 play positive roles in the expression of CD49f. Indeed, CD49f expression was upregulated in human embryonic stem cells (hESCs) compared with hMSCs. The elevated level of CD49f expression was significantly decreased upon embryoid body formation in hESCs. In hESCs, the knockdown of CD49f downregulated PI3K/AKT signaling and upregulated the level of p53, inducing differentiation into three germ layers. Taken together, our data suggest that the cell-surface protein CD49f has novel and dynamic roles in regulating the differentiation potential of hMSCs and maintaining pluripotency.


Asunto(s)
Células Madre Embrionarias/metabolismo , Integrina alfa6/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Factores de Transcripción SOXB1/biosíntesis , Diferenciación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Regulación de la Expresión Génica/fisiología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/metabolismo
7.
Elife ; 122023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36880644

RESUMEN

Stem cell differentiation requires dramatic changes in gene expression and global remodeling of chromatin architecture. How and when chromatin remodels relative to the transcriptional, behavioral, and morphological changes during differentiation remain unclear, particularly in an intact tissue context. Here, we develop a quantitative pipeline which leverages fluorescently-tagged histones and longitudinal imaging to track large-scale chromatin compaction changes within individual cells in a live mouse. Applying this pipeline to epidermal stem cells, we reveal that cell-to-cell chromatin compaction heterogeneity within the stem cell compartment emerges independent of cell cycle status, and instead is reflective of differentiation status. Chromatin compaction state gradually transitions over days as differentiating cells exit the stem cell compartment. Moreover, establishing live imaging of Keratin-10 (K10) nascent RNA, which marks the onset of stem cell differentiation, we find that Keratin-10 transcription is highly dynamic and largely precedes the global chromatin compaction changes associated with differentiation. Together, these analyses reveal that stem cell differentiation involves dynamic transcriptional states and gradual chromatin rearrangement.


Asunto(s)
Cromatina , Queratina-10 , Animales , Ratones , Queratina-10/genética , Queratina-10/metabolismo , Histonas/metabolismo , Diferenciación Celular/genética , Células Madre/metabolismo
8.
Nat Commun ; 14(1): 8245, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38086920

RESUMEN

Pluripotent stem cell-derived organoids can recapitulate significant features of organ development in vitro. We hypothesized that creating human heart organoids by mimicking aspects of in utero gestation (e.g., addition of metabolic and hormonal factors) would lead to higher physiological and anatomical relevance. We find that heart organoids produced using this self-organization-driven developmental induction strategy are remarkably similar transcriptionally and morphologically to age-matched human embryonic hearts. We also show that they recapitulate several aspects of cardiac development, including large atrial and ventricular chambers, proepicardial organ formation, and retinoic acid-mediated anterior-posterior patterning, mimicking the developmental processes found in the post-heart tube stage primitive heart. Moreover, we provide proof-of-concept demonstration of the value of this system for disease modeling by exploring the effects of ondansetron, a drug administered to pregnant women and associated with congenital heart defects. These findings constitute a significant technical advance for synthetic heart development and provide a powerful tool for cardiac disease modeling.


Asunto(s)
Cardiopatías , Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Embarazo , Humanos , Femenino , Células Madre Pluripotentes Inducidas/metabolismo , Organoides/metabolismo , Corazón , Cardiopatías/metabolismo , Diferenciación Celular/fisiología
9.
bioRxiv ; 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-37986907

RESUMEN

Hematophagous ectoparasites, such as ticks, rely on impaired wound healing for skin attachment and blood feeding. Wound healing has been extensively studied through the lens of inflammatory disorders and cancer, but limited attention has been given to arthropod-borne diseases. Here, we used orthogonal approaches combining single-cell RNA sequencing (scRNAseq), flow cytometry, murine genetics, and intravital microscopy to demonstrate how tick extracellular vesicles (EVs) disrupt networks involved in tissue repair. Impairment of EVs through silencing of the SNARE protein vamp33 negatively impacted ectoparasite feeding and survival in three medically relevant tick species, including Ixodes scapularis. Furthermore, I. scapularis EVs affected epidermal γδ T cell frequencies and co-receptor expression, which are essential for keratinocyte function. ScRNAseq analysis of the skin epidermis in wildtype animals exposed to vamp33-deficient ticks revealed a unique cluster of keratinocytes with an overrepresentation of pathways connected to wound healing. This biological circuit was further implicated in arthropod fitness when tick EVs inhibited epithelial proliferation through the disruption of phosphoinositide 3-kinase activity and keratinocyte growth factor levels. Collectively, we uncovered a tick-targeted impairment of tissue repair via the resident γδ T cell-keratinocyte axis, which contributes to ectoparasite feeding.

10.
Cytotherapy ; 14(5): 630-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22404083

RESUMEN

BACKGROUND AIMS: Niemann-Pick disease type C1 (NPC) is an autosomal recessive cholesterol-storage disorder characterized by liver dysfunction, hepatosplenomegaly and progressive neurodegeneration. Thus far, studies of NPC mice have been performed mainly to study the brain and neurodegeneration, because degeneration in the brain was known as the primary cause of death in NPC mice. However, NPC is a systemic disease; therefore the purpose of this study was to find the possibility of a general therapeutic effect by applying and tracking transplanted human amniotic epithelial stem cells (hAESC) in NPC mice. METHODS: hAESC were administered to NPC homozygous (NPC(-/-)) mice via intravenous injection from 5 weeks of age; each recipient received 5 × 10(5) cells every other week. The body weight of each of the mice was measured every week, and the survival and state of each mouse was evaluated every day. The weight of the organs was measured, and serum chemistry, histology and the intensity of Filipin staining were evaluated. RESULTS: The effect of cell transplantation was to extend the life span and reduce the rapid loss of weight. Moreover, alleviation of tissue damage was observed more in hAESC-treated NPC(-/-) mice than in non-treated NPC(-/-) mice. Cholesterol deposition was reduced after transplantation, and the relative weight of the liver was also decreased. CONCLUSIONS: These data show that hAESC could delay the degeneration caused by fatal genetic disorders such as NPC. This study presents the prospect of relief of precipitous disease progression and the therapeutic possibility of applying hAESC to fatal genetic disorders.


Asunto(s)
Amnios/citología , Células Epiteliales/citología , Enfermedad de Niemann-Pick Tipo C/terapia , Trasplante de Células Madre , Células Madre/citología , Amnios/metabolismo , Animales , Colesterol/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Humanos , Inyecciones Intravenosas , Hepatopatías/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/patología , Insuficiencia Renal/terapia , Células Madre/metabolismo
11.
Cell Mol Life Sci ; 68(2): 325-36, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20652617

RESUMEN

Cellular senescence involves a reduction in adult stem cell self-renewal, and epigenetic regulation of gene expression is one of the main underlying mechanisms. Here, we observed that the cellular senescence of human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs) caused by inhibition of histone deacetylase (HDAC) activity leads to down-regulation of high mobility group A2 (HMGA2) and, on the contrary, to up-regulation of p16(INK)4(A), p21(CIP)¹(/WAF)¹ and p27(KIP)¹. We found that let-7a1, let-7d, let-7f1, miR-23a, miR-26a and miR-30a were increased during replicative and HDAC inhibitor-mediated senescence of hUCB-MSCs by microRNA microarray and real-time quantitative PCR. Furthermore, the configurations of chromatins beading on these miRNAs were prone to transcriptional activation during HDAC inhibitor-mediated senescence. We confirmed that miR-23a, miR-26a and miR-30a inhibit HMGA2 to accelerate the progress of senescence. These findings suggest that HDACs may play important roles in cellular senescence by regulating the expression of miRNAs that target HMGA2 through histone modification.


Asunto(s)
Células Madre Adultas/metabolismo , Senescencia Celular/fisiología , Proteínas del Grupo de Alta Movilidad , Histona Desacetilasas , MicroARNs/metabolismo , Células Madre Multipotentes/metabolismo , Tejido Adiposo/citología , Adulto , Células Madre Adultas/citología , Proliferación Celular , Activación Enzimática/efectos de los fármacos , Epigenómica , Femenino , Sangre Fetal/citología , Sangre Fetal/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , MicroARNs/genética , MicroARNs/farmacología , Análisis por Micromatrices , Células Madre Multipotentes/citología , Reacción en Cadena de la Polimerasa , Ácido Valproico/farmacología
12.
Front Cell Dev Biol ; 10: 933370, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35938157

RESUMEN

Hair follicles are mini organs that repeat the growth and regression cycle continuously. These dynamic changes are driven by the regulation of stem cells via their multiple niche components. To build the complex structure of hair follicles and surrounding niches, sophisticated morphogenesis is required during embryonic development. This review will explore how hair follicles are formed and maintained through dynamic cellular changes and diverse signaling pathways. In addition, comparison of differences in stem cells and surrounding niche components during embryogenesis, neogenesis, and organogenesis will provide a comprehensive understanding of mechanisms for hair follicle generation and insights into skin regeneration.

13.
Cytotherapy ; 13(3): 341-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20860427

RESUMEN

BACKGROUND AIMS: Amniotic fluid (AF) is a well-known source of stem cells. However, there have been no reports regarding equine AF stem cells. We have isolated equine AF-derived multipotent stem cells (MSC) (eAF-MSC) and show that these cells exhibit self-renewal ability and multilineage differentiation. METHODS: AF was obtained from thoroughbred mares and mononuclear cells (MNC) were isolated by Ficoll-Paque density gradient. We measured the cumulative population doubling level (CPDL) and characterized the immunophenotype by flow cytometry. To investigate differentiation ability, a trilineage differentiation assay was conducted. RESULTS: eAF-MSC could be isolated and the proliferation level was high. eAF-MSC presented typical MSC phenotypic markers, as determined by flow cytometry. Moreover, eAF-MSC showed a trilineage differentiation capability. CONCLUSIONS: Equine AF is a good source of MSC. Furthermore, eAF-MSC may be useful as a cell therapy application for horses.


Asunto(s)
Líquido Amniótico/citología , Separación Celular/métodos , Células Madre Multipotentes/citología , Adipogénesis , Animales , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Condrogénesis , Citometría de Flujo , Caballos , Humanos , Inmunofenotipificación , Células Madre Multipotentes/metabolismo , Osteogénesis , Homología de Secuencia de Aminoácido
14.
Cell Mol Life Sci ; 67(7): 1165-76, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20049504

RESUMEN

Aging is linked to loss of the self-renewal capacity of adult stem cells. Here, we observed that human multipotent stem cells (MSCs) underwent cellular senescence in vitro. Decreased expression of histone deacetylases (HDACs), followed by downregulation of polycomb group genes (PcGs), such as BMI1, EZH2 and SUZ12, and by upregulation of jumonji domain containing 3 (JMJD3), was observed in senescent MSCs. Similarly, HDAC inhibitors induced cellular senescence through downregulation of PcGs and upregulation of JMJD3. Regulation of PcGs was associated with HDAC inhibitor-induced hypophosphorylation of RB, which causes RB to bind to and decrease the transcriptional activity of E2F. JMJD3 expression regulation was dependant on histone acetylation status at its promoter regions. A histone acetyltransferase (HAT) inhibitor prevented replicative senescence of MSCs. These results suggest that HDAC activity might be important for MSC self-renewal by balancing PcGs and JMJD3 expression, which govern cellular senescence by p16(INK4A) regulation.


Asunto(s)
Células Madre Adultas/metabolismo , Senescencia Celular , Histona Desacetilasas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células Madre Multipotentes/metabolismo , Proteínas Represoras/metabolismo , Células Madre Adultas/citología , Células Cultivadas , Regulación hacia Abajo , Epigénesis Genética , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Células Madre Multipotentes/citología , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/genética , Regulación hacia Arriba , Ácido Valproico/farmacología
15.
Front Cell Dev Biol ; 9: 796080, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35145968

RESUMEN

Wound repair is essential to restore tissue function through the rebuilding of pre-existing structures. The repair process involves the re-formation of tissue, which was originally generated by embryonic development, with as similar a structure as possible. Therefore, these two processes share many similarities in terms of creating tissue architecture. However, fundamental differences still exist, such as differences in the cellular components, the status of neighboring tissues, and the surrounding environment. Recent advances in single-cell transcriptomics, in vivo lineage tracing, and intravital imaging revealed subpopulations, long-term cell fates, and dynamic cellular behaviors in live animals that were not detectable previously. This review highlights similarities and differences between adult wound repair and embryonic tissue development with a particular emphasis on the epidermis of the skin.

16.
Vet Sci ; 8(9)2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34564576

RESUMEN

Adipose tissue-derived mesenchymal stem cells (AD-MSCs) release extracellular vesicles such as exosomes, apoptotic bodies, and microparticles. In particular, exosomes are formed inside cells via multivesicular bodies (MVBs), thus their protein, DNA, and RNA content are similar to those of the parent cells. Exosome research is rapidly expanding, with an increase in the number of related publications observed in recent years; therefore, the function and application of MSC-derived exosomes could emerge as cell-free therapeutics. Exosomes have been isolated from feline AD-MSCs and feline fibroblast cell culture media using ultracentrifugation. Feline exosomes have been characterized by FACS, nanoparticle tracking analysis, and transmission electron microscopy imaging. Moreover, cytokine levels were detected by sandwich enzyme-linked immunosorbent assay in exosomes and LPS-induced THP-1 macrophages. The size of the isolated exosomes was that of a typical exosome, i.e., approximately 150 nm, and they expressed tetraspanins CD9 and CD81. The anti-inflammatory factor IL-10 was increased in feline AD-MSC-derived exosomes. However, pro-inflammatory factors such as IL-1ß, IL-8, IL-2, RANTES, and IFN-gamma were significantly decreased in feline AD-MSC-derived exosomes. This was the first demonstration that feline AD-MSC-derived exosomes enhance the inflammatory suppressive effects and have potential for the treatment of immune diseases or as an inflammation-inhibition therapy.

17.
Nat Cell Biol ; 23(5): 476-484, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33958758

RESUMEN

Organs consist of multiple cell types that ensure proper architecture and function. How different cell types coexist and interact to maintain their homeostasis in vivo remains elusive. The skin epidermis comprises mostly epithelial cells, but also harbours Langerhans cells (LCs) and dendritic epidermal T cells (DETCs). Whether and how distributions of LCs and DETCs are regulated during homeostasis is unclear. Here, by tracking individual cells in the skin of live adult mice over time, we show that LCs and DETCs actively maintain a non-random spatial distribution despite continuous turnover of neighbouring basal epithelial cells. Moreover, the density of epithelial cells regulates the composition of LCs and DETCs in the epidermis. Finally, LCs require the GTPase Rac1 to maintain their positional stability, density and tiling pattern reminiscent of neuronal self-avoidance. We propose that these cellular mechanisms provide the epidermis with an optimal response to environmental insults.


Asunto(s)
Células Epidérmicas/citología , Epidermis/metabolismo , Piel/citología , Linfocitos T/inmunología , Animales , Células Epidérmicas/inmunología , Epidermis/inmunología , Homeostasis/inmunología , Homeostasis/fisiología , Uniones Intercelulares/patología , Ratones Transgénicos , Piel/inmunología
18.
Ann N Y Acad Sci ; 1462(1): 27-36, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31655007

RESUMEN

Adult stem cells are rare, undifferentiated cells found in all tissues of the body. Although normally kept in a quiescent, nondividing state, these cells can proliferate and differentiate to replace naturally dying cells within their tissue and to repair its wounds in response to injury. Due to their proliferative nature and ability to regenerate tissue, adult stem cells have the potential to treat a variety of degenerative diseases as well as aging. In addition, since stem cells are often thought to be the source of malignant tumors, understanding the mechanisms that keep their proliferative abilities in check can pave the way for new cancer therapies. While adult stem cells have had limited practical and clinical applications to date, several clinical trials of stem cell-based therapies are underway. This report details recent research presented at the New York Academy of Sciences on March 14, 2019 on understanding the factors that regulate stem cell activity and differentiation, with the hope of translating these findings into the clinic.


Asunto(s)
Células Madre Adultas/trasplante , Medicina Regenerativa/tendencias , Informe de Investigación/tendencias , Trasplante de Células Madre/tendencias , Adulto , Células Madre Adultas/fisiología , Envejecimiento/patología , Animales , Diferenciación Celular/fisiología , Humanos , Neoplasias/patología , Neoplasias/terapia , Ciudad de Nueva York , Medicina Regenerativa/métodos , Trasplante de Células Madre/métodos
19.
Growth Factors ; 27(6): 425-37, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19919531

RESUMEN

It has widely been reported that basic fibroblast growth factor (bFGF) promotes proliferation of human stem cells and contributes to the maintenance of their self-renewal capability through repeated replications. In contrast to embryonic stem cells (ESCs), the effects of growth factors on adult stem cells are poorly understood. In human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs), bFGF is associated with an increased number of proliferating cells. Furthermore, expression levels of ESC markers were increased after treatment with bFGF. bFGF also increased the expression of FGFR, which in turn increased expression of insulin-like growth factor (IGFs). Since IGFs exert autocrine and paracrine effects on stem cells, bFGF-mediated release of IGFs from hUCB-MSCs might enhance FGFR1 and IGF1R expression in neighboring cells. These receptors could subsequently regulate the effects of bFGF and IGFs in adult stem cells. These results suggest that positive feedback regulation of bFGF and IGFs leads to proliferation of hUCB-MSCs.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica , Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre Multipotentes/efectos de los fármacos , Adulto , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Femenino , Sangre Fetal , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/genética , Células Madre Multipotentes/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
20.
Biochem Biophys Res Commun ; 384(1): 120-5, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19394308

RESUMEN

The OCT4A gene, a POU homeodomain transcription factor, has been shown to be expressed in embryonic stem cells (ESC) as well as hUCB-MSCs. In this study, the roles played by OCT4A in hUCB-MSCs were determined by stably inhibiting OCT4A with lenti-viral vector-based small hairpin RNA (shRNA). A decreased rate of cell proliferation was observed in OCT4-inhibited hUCB-MSCs. Down-regulation of CCNA2 expression in OCT4-inhibited hUCB-MSCs was confirmed by RT-PCR and real-time RT-PCR analysis in three genetically independent hUCB-MSC clones. Adipogenic differentiation was also suppressed in OCT4-inhibited hUCB-MSCs. The up-regulation of DTX1 and down-regulation of HDAC1, 2, and 4 expressions may be related to this differentiation deformity. The expression of other transcription factors, including SOX2, REX1 and c-MYC, was also affected by OCT4 inhibition in hUCB-MSCs. In conclusion, these finding suggest that OCT4A performs functionally conserved roles in hUCB-MSCs, making its expression biologically important for ex vivo culture of hUCB-MSCs.


Asunto(s)
Sangre Fetal/citología , Células Madre Multipotentes/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Adipogénesis/genética , Ciclo Celular/genética , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Expresión Génica , Humanos , Células Madre Multipotentes/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA