Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Perinatol ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38688998

RESUMEN

OBJECTIVE: Intraventricular hemorrhage (IVH) is a common cause of preterm brain injury. Fresh parent's own milk (POM) contains pluripotent stem cells (SCs) that produce neuronal cells in-vitro. The permeable neonatal blood brain barrier potentially allows SC delivery. We performed the first prospective trial (clinicaltrials.gov NCT04225286) of feasibility of intranasal POM (IPOM) in preterm infants with IVH and described SC content of POM samples. STUDY DESIGN: 37 Infants (mean gestation 27.7 ± 2.6 weeks, birthweight 1030 ± 320 g) with IVH (35.1% grade IV) were recruited from two tertiary Toronto NICUs. IPOM was given ideally twice daily until 28 days of age. Tolerance and adverse reactions were collected and 162 administering providers surveyed. RESULTS: There were no major adverse reactions. Provider surveys suggested acceptability, although potential provider and subject stress requires further study. Milk cell analysis suggests wide variability between parents. CONCLUSIONS: This phase 1 study demonstrated IPOM was tolerated and feasible in preterm infants.

2.
NPJ Regen Med ; 8(1): 45, 2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37626067

RESUMEN

Cell-based therapeutics are promising interventions to repair ischemic cardiac tissue. However, no single cell type has yet been found to be both specialized and versatile enough to heal the heart. The synergistic effects of two regenerative cell types including endothelial colony forming cells (ECFC) and first-trimester human umbilical cord perivascular cells (FTM HUCPVC) with endothelial cell and pericyte properties respectively, on angiogenic and regenerative properties were tested in a rat model of myocardial infarction (MI), in vitro tube formation and Matrigel plug assay. The combination of FTM HUCPVCs and ECFCs synergistically reduced fibrosis and cardiomyocyte apoptosis, while promoting favorable cardiac remodeling and contractility. These effects were in part mediated by ANGPT2, PDGF-ß, and VEGF-C. PDGF-ß signaling-dependent synergistic effects on angiogenesis were also observed in vitro and in vivo. FTM HUCPVCs and ECFCs represent a cell combination therapy for promoting and sustaining vascularization following ischemic cardiac injury.

3.
J Inflamm (Lond) ; 20(1): 15, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37127610

RESUMEN

BACKGROUND: The Kynurenine Pathway (KP) of tryptophan degradation and glutamate toxicity is implicated in several neurological disorders, including depression. The therapeutic potential of mesenchymal stromal cells (MSC), owing to their well documented phagocytosis-driven mechanism of immunomodulation and neuroprotection, has been tested in many neurological disorders. However, their potential to influence KP and the glutamatergic system has not yet been investigated. Hence, this study sought to investigate the effect of HUCPVC, a rich and potent source of MSC, on Lipopolysaccharide (LPS)-activated KP metabolites, KP enzymes, and key components of glutamate neurotransmission. METHODS: The immunomodulatory effect of peripherally administered HUCPVC on the expression profile of kynurenine pathway metabolites and enzymes was assessed in the plasma and brain of mice treated with LPS using LCMS and QPCR. An assessment of the glutamatergic system, including selected receptors, transporters and related proteins was also conducted by QPCR, immunohistochemistry and Western blot. RESULTS: HUCPVC were found to modulate LPS-induced activation of KP enzymes and metabolites in the brain associated with neurotoxicity. Moreover, the reduced expression of the glutamatergic components due to LPS was also found to be significantly improved by HUCPVC. CONCLUSIONS: The immunomodulatory properties of HUCPVC appear to confer neuroprotection, at least in part, through their ability to modulate the KP in the brain. This KP modulation enhances neuroprotective regulators and downregulates neurotoxic consequences, including glutamate neurotoxicity, which is associated with neuroinflammation and depressive behavior.

4.
Mol Cell Proteomics ; 22(6): 100556, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37087050

RESUMEN

Non-obstructive azoospermia (NOA), the most severe form of male infertility, could be treated with intracytoplasmic sperm injection, providing spermatozoa were retrieved with the microdissection testicular sperm extraction (mTESE). We hypothesized that testis-specific and germ cell-specific proteins would facilitate flow cytometry-assisted identification of rare spermatozoa in semen cell pellets of NOA patients, thus enabling non-invasive diagnostics prior to mTESE. Data mining, targeted proteomics, and immunofluorescent microscopy identified and verified a panel of highly testis-specific proteins expressed at the continuum of germ cell differentiation. Late germ cell-specific proteins AKAP4_HUMAN and ASPX_HUMAN (ACRV1 gene) revealed exclusive localization in spermatozoa tails and acrosomes, respectively. A multiplex imaging flow cytometry assay facilitated fast and unambiguous identification of rare but morphologically intact AKAP4+/ASPX+/Hoechst+ spermatozoa within debris-laden semen pellets of NOA patients. While the previously suggested markers for spermatozoa retrieval suffered from low diagnostic specificity, the multistep gating strategy and visualization of AKAP4+/ASPX+/Hoechst+ cells with elongated tails and acrosome-capped nuclei facilitated fast and unambiguous identification of the mature intact spermatozoa. AKAP4+/ASPX+/Hoechst+ assay may emerge as a noninvasive test to predict retrieval of morphologically intact spermatozoa by mTESE, thus improving diagnostics and treatment of severe forms of male infertility.


Asunto(s)
Azoospermia , Infertilidad Masculina , Masculino , Humanos , Azoospermia/genética , Azoospermia/metabolismo , Azoospermia/terapia , Semen/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Infertilidad Masculina/metabolismo , Estudios Retrospectivos , Proteínas de Anclaje a la Quinasa A/metabolismo
5.
Cytotherapy ; 25(2): 125-137, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36473795

RESUMEN

BACKGROUND AIMS: Because of their potent immunomodulatory and anti-inflammatory properties, mesenchymal stromal cells are a major focus in the field of stem cell therapy. However, the precise mechanisms underlying this are not entirely understood. Human umbilical cord perivascular cells (HUCPVCs) are a promising cell therapy candidate. This study was designed to evaluate the time course and mechanisms by which HUCPVCs mitigate lipopolysaccharide (LPS)-induced systemic and neurological inflammation in immunocompetent mice. To explore the underlying mechanisms, the authors investigated the biodistribution and fate of HUCPVCs. METHODS: Male C57BL/6 mice were randomly allocated to four groups: control, LPS, HUCPVCs or LPS + HUCPVCs. Quantitative polymerase chain reaction, enzyme-linked immunosorbent assay and cytokine arrays were used to assess changes in pro-inflammatory mediators systemically and in the brain. Depressive-like behavioral changes were evaluated via a forced swim test. Quantum dot (qDot) labeling and immunohistochemistry were used to assess the biodistribution and fate of HUCPVCs and interactions with recipient innate immune cells. RESULTS: A single intravenously delivered dose of HUCPVCs significantly reduced the systemic inflammation induced by LPS within the first 24 h after administration. HUCPVC treatment abrogated the upregulated expression of pro-inflammatory genes in the hippocampus and cortex and attenuated depressive-like behavior induced by LPS treatment. Biodistribution analysis revealed that HUCPVC-derived qDots rapidly accumulated in the lungs and demonstrated limited in vivo persistence. Furthermore, qDot signals were associated with major recipient innate immune cells and promoted a shift in macrophages toward a regulatory phenotype in the lungs. CONCLUSIONS: Overall, this study demonstrates that HUCPVCs can successfully reduce systemic and neurological inflammation induced by LPS within the first 24 h after administration. Biodistribution and fate analyses suggest a critical role for the innate immune system in the HUCPVC-based immunomodulation mechanism.


Asunto(s)
Lipopolisacáridos , Células Madre Mesenquimatosas , Animales , Masculino , Ratones , Inflamación/inducido químicamente , Inflamación/terapia , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Ratones Endogámicos C57BL , Distribución Tisular , Cordón Umbilical , Células Endoteliales de la Vena Umbilical Humana , Humanos
6.
Cancer Lett ; 501: 133-146, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33387641

RESUMEN

Chemotherapies can cause germ cell depletion and gonadal failure. When injected post-chemotherapy, mesenchymal stromal cells (MSCs) from various sources have been shown to have regenerative effects in rodent models of chemotherapy-induced gonadal injury. Here, we evaluated two properties of a novel source of MSC, first trimester (FTM) human umbilical cord perivascular cells (HUCPVCs) (with increased regenerative potential compared to older sources), that may render them a promising candidate for chemotherapeutic gonadal injury prevention. Firstly, their ability to resist the cytotoxic effects of cyclophosphamide (CTX) in vitro, as compared to term HUCPVCs and bone marrow cells (BMSCs); and secondly, whether they prevent gonadal dysfunction if delivered prior to gonadotoxic therapy in vivo. BMSC, FTM HUCPVC, term HUCPVC, and control NTERA2 cells were treated with moderate (150 µmol/L) and high (300 µmol/L) doses of CTX in vitro. Viability, proliferative capacity, mesenchymal cell lineage markers and differentiation capacity, immunogenicity, and paracrine gene expression were assessed. CTX was administered to Wistar rats 2 days following an intra-ovarian injection of FTM HUCPVC. HUCPVC survival and ovarian follicle numbers were assessed using histological methods. We conclude that FTM HUCPVC maintain key regenerative properties following chemotherapy exposure and that pre-treatment with these cells may prevent CTX-induced ovarian damage in vivo. Therefore, HUCPVCs are promising candidates for fertility preservation.


Asunto(s)
Ciclofosfamida/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Regeneración/fisiología , Cordón Umbilical/citología , Cordón Umbilical/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ciclofosfamida/efectos adversos , Relación Dosis-Respuesta a Droga , Femenino , Preservación de la Fertilidad , Humanos , Ovario/efectos de los fármacos , Ratas , Ratas Wistar , Regeneración/efectos de los fármacos , Cordón Umbilical/trasplante
7.
F S Sci ; 2(1): 24-32, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-35559762

RESUMEN

OBJECTIVE: To study whether intratesticular (IT) administration of 2 sources of human umbilical cord perivascular cells (HUCPVC), rich and potent sources of mesenchymal stromal cells (MSC), before chemotherapy can prevent infertility in a mouse model. DESIGN: Two control groups of CD1 male mice without busulfan (BUS) administration (untreated and IT media injection groups) were included. Experimental groups included IT administration of media, first trimester (FTM) HUCPVCs or term HUCPVCs (n = 5 each) injected 3 days before BUS treatment (20 mg/kg). All groups were included in a mating time course study over 6 months. SETTING: Preclinical study in a fertility center research laboratory. PATIENTS: Not applicable. INTERVENTION: IT delivery of FTM or term HUCPVC before BUS treatment. MAIN OUTCOME MEASURES: Pregnancies, litter sizes, and gross morphology of offspring were monitored. Caudal epididymal sperm concentration, motility, and progressive motility were assessed by computer-assisted sperm analysis. Spermatogenesis was also assessed histologically in testicular tissue sections. RESULTS: FTM and term HUCPVC displayed an MSC-associated immunophenotype and expressed transcripts encoding paracrine factors known to regulate the testicular cell niche. IT administration of FTM and term HUCPVC before chemotherapy promoted the recovery of spermatogenesis and fertility compared with BUS-treated animals that received a media injection. Although the total number of pups sired over 6 months by males treated with FTM or term HUCPVC was reduced compared with untreated or media-injected controls, litter size and sperm parameters in fertile animals did not differ between control and cell-treated groups. CONCLUSION: HUCPVC represent a promising source of MSC-based therapy to prevent gonadotoxic chemotherapeutic drug-induced infertility.


Asunto(s)
Infertilidad Masculina , Células Madre Mesenquimatosas , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Infertilidad Masculina/inducido químicamente , Masculino , Ratones , Embarazo , Espermatogénesis , Cordón Umbilical/irrigación sanguínea
8.
Stem Cells Dev ; 29(4): 198-211, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31701812

RESUMEN

Traumatic brain injury (TBI) leads to delayed secondary injury events consisting of cellular and molecular cascades that exacerbate the initial injury. Human umbilical cord perivascular cells (HUCPVCs) secrete neurotrophic and prosurvival factors. In this study, we examined the effects of HUCPVC in sympathetic axon and cortical axon survival models and sought to determine whether HUCPVC provide axonal survival cues. We then examined the effects of the HUCPVC in an in vivo fluid percussion injury model of TBI. Our data indicate that HUCPVCs express neurotrophic and neural survival factors. They also express and secrete relevant growth and survival proteins when cultured alone, or in the presence of injured axons. Coculture experiments indicate that HUCPVCs interact preferentially with axons when cocultured with sympathetic neurons and reduce axonal degeneration. Nerve growth factor withdrawal in axonal compartments resulted in 66 ± 3% axon degeneration, whereas HUCPVC coculture rescued axon degeneration to 35 ± 3%. Inhibition of Akt (LY294002) resulted in a significant increase in degeneration compared with HUCPVC cocultures (48 ± 7% degeneration). Under normoxic conditions, control cultures showed 39 ± 5% degeneration. Oxygen glucose deprivation (OGD) resulted in 58 ± 3% degeneration and OGD HUCPVC cocultures reduced degeneration to 34 ± 5% (p < 0.05). In an in vivo model of TBI, immunohistochemical analysis of NF200 showed improved axon morphology in HUCPVC-treated animals compared with injured animals. These data presented in this study indicate an important role for perivascular cells in protecting axons from injury and a potential cell-based therapy to treat secondary injury after TBI.


Asunto(s)
Axones/metabolismo , Lesiones Traumáticas del Encéfalo/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Neuronas/metabolismo , Pericitos/trasplante , Animales , Axones/efectos de los fármacos , Axones/patología , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/patología , Cromonas/farmacología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Glucosa/deficiencia , Glucosa/farmacología , Humanos , Morfolinas/farmacología , Factor de Crecimiento Nervioso/farmacología , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Oxígeno/farmacología , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/metabolismo , Cordón Umbilical/citología , Cordón Umbilical/metabolismo
9.
Biol Psychiatry ; 86(9): 712-724, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31521333

RESUMEN

BACKGROUND: Hyperactivation of innate immunity has been implicated in the etiology of mood disorders, including major depressive disorder (MDD). Mesenchymal stromal cells (MSCs) have demonstrated potent immunomodulatory capabilities in the context of chronic inflammatory disease and injury but have yet to be evaluated in stress-based preclinical models of MDD. We sought to test the ability of intravenous MSCs to modulate innate immune activation and behavioral patterns associated with repeated social defeat (RSD). METHODS: Murine RSD-induced innate immune activation as well as depressive and anxiety-like behaviors were assessed in unstressed, RSD, and RSD + human MSC groups. Biodistribution and fate studies were performed to inform potential mechanisms of action. RESULTS: MSCs reduced stress-induced circulating proinflammatory cytokines, monocytes, neuroinflammation, and depressive and anxiety-like behaviors. Biodistribution analyses indicated that infused MSCs distributed within peripheral organs without homing to the brain. Murine neutrophils targeted MSCs in the lungs within hours of administration. MSCs and recipient neutrophils were cleared by recipient macrophages promoting a switch toward a regulatory phenotype and systemic resolution of inflammation. CONCLUSIONS: Peripheral delivery of MSCs modulates central nervous system inflammatory processes and aberrant behavioral patterns in a stress-based rodent model of MDD and anxiety. Recent studies suggest that host immune cell-mediated phagocytosis of MSCs in vivo can trigger an immunomodulatory cascade, resulting in resolution of inflammation. Our data suggest that similar mechanisms may protect distal organs, including the brain, from systemic, stress-induced proinflammatory spikes and may uncover unexpected targets in the periphery for novel or adjunct treatment for a subset of patients with MDD.


Asunto(s)
Conducta , Proliferación Celular , Inflamación/inmunología , Macrófagos/inmunología , Células Madre Mesenquimatosas/inmunología , Animales , Ansiedad/inmunología , Biomarcadores , Células Cultivadas , Citocinas/inmunología , Depresión/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Ratones , Monocitos/inmunología , Distribución Tisular
10.
Adv Exp Med Biol ; 1122: 211-233, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30937871

RESUMEN

The structural components of the umbilical cord, including two arteries and one vein, the stromal region/Wharton's jelly, and amniotic epithelial membrane, are well described at various time points of gestation. Over the last two decades, evidence has emerged that multipotent cells sharing properties of mesenchymal stromal cell and pericytes/mural cells can be isolated from multiple regions of the umbilical cord, including the perivascular region of the umbilical cord arteries and vein, Wharton's jelly, and subamnion. These cells have increasingly gained interest for their potential use in regenerative and immunomodulatory medicine. Recent studies suggest that obstetrical complications including gestational diabetes mellitus and preeclampsia may alter the yield, properties, and potency of mesenchymal stromal cells isolated from the umbilical cord. The role that pericytes or pericyte-like cells play in the development of the human umbilical cord and associated pathologies, however, remains to be investigated.


Asunto(s)
Células Madre Mesenquimatosas/citología , Pericitos/citología , Cordón Umbilical/citología , Amnios/citología , Diferenciación Celular , Femenino , Humanos , Embarazo , Complicaciones del Embarazo/patología , Gelatina de Wharton/citología
11.
Stem Cell Res Ther ; 8(1): 184, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28807010

RESUMEN

BACKGROUND: Due to limitations of current angiogenesis assays, we aimed to develop a novel application of the rat aortic ring assay to assess the angiogenic potential of mesenchymal stromal cells (MSCs). First-trimester human umbilical cord-derived perivascular cells (FTM HUCPVCs) have multipotent characteristics and previously demonstrated angiogenic potential. We compared the effect of this young source of MSCs and adult bone marrow stromal cells (BMSCs) on ex vivo aortic endothelial network formation. METHODS: Thoracic segments of adult rat aortas were isolated, sectioned and embedded into Matrigel™. Fluorophore-labeled FTM HUCPVC lines and BMSCs (N = 3) were cocultured with developing endothelial networks (day 0). MSC integration, tube formation and endothelial network growth were monitored daily using phase-contrast and fluorescence microscopy. Quantification of endothelial networks was performed using ImageJ network analysis software on day 5 of coculture. RESULTS: FTM HUCPVCs from two umbilical cord samples migrated toward and integrated with developing aortic ring tubular networks while displaying elongated morphologies (day 1). In contrast, BMSCs did not show targeted migration and maintained spherical morphologies with limited physical interactions. Within 1 week of coculture, FTM HUCPVC lines contributed to significantly greater radial network growth and network loop formation when compared to BMSCs and untreated networks. CONCLUSIONS: We have developed a novel potency assay to assess the angiogenic potential of cell therapy candidates. Favorable properties of FTM HUCPVCs over BMSCs that we observed with this assay and which merit further study include chemotaxis, affinity for developing vasculature, and physical supportive interactions contributing to the development of endothelial networks.


Asunto(s)
Aorta/fisiología , Bioensayo/métodos , Tratamiento Basado en Trasplante de Células y Tejidos , Neovascularización Fisiológica , Animales , Movimiento Celular , Forma de la Célula , Técnicas de Cocultivo , Células Endoteliales/citología , Femenino , Humanos , Técnicas In Vitro , Células Madre Mesenquimatosas/citología , Microscopía Fluorescente , Pericitos/citología , Ratas Sprague-Dawley , Cordón Umbilical/citología
12.
Fertil Steril ; 107(3): 595-605.e7, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28259258

RESUMEN

OBJECTIVE: To optimize culture conditions for human testicular somatic cells (TSCs) and spermatogonial stem cells. DESIGN: Basic science study. SETTING: Urology clinic and stem cell research laboratory. PATIENT(S): Eight human testicular samples. INTERVENTIONS(S): Testicular tissues were processed by mechanical and enzymatic digestion. Cell suspensions were subjected to differential plating (DP) after which floating cells (representing germ cells) were removed and attached cells (representing TSCs) were cultured for 2 passages (P0-P1) in StemPro-34- or DMEM-F12-based medium. Germ cell cultures were established in both media for 12 days. MAIN OUTCOME MEASURE(S): TSC cultures: proliferation doubling time (PDT), fluorescence-activated cell sorting for CD90, next-generation sequencing for 89 RNA transcripts, immunocytochemistry for TSC and germ cell markers, and conditioned media analysis; germ cell cultures: number of aggregates. RESULT(S): TSCs had significantly prolonged PDT in DMEM-F12 versus StemPro-34 (319.6 ± 275.8 h and 110.5 ± 68.3 h, respectively). The proportion of CD90-positive cells increased after P1 in StemPro-34 and DMEM-F12 (90.1 ± 10.8% and 76.5 ± 17.4%, respectively) versus after DP (66.3 ± 7%). Samples from both media after P1 clustered closely in the principle components analysis map whereas those after DP did not. After P1 in either medium, CD90-positive cells expressed TSC markers only, and fibroblast growth factor 2 and bone morphogenetic protein 4 were detected in conditioned medium. A higher number of germ cell aggregates formed in DMEM-F12 (59 ± 39 vs. 28 ± 17, respectively). CONCLUSION(S): Use of DMEM-F12 reduces TSC proliferation while preserving their unique characteristics, leading to improved germ cell aggregates formation compared with StemPro-34, the standard basal medium used in the majority of previous reports.


Asunto(s)
Células Madre Germinales Adultas/fisiología , Proliferación Celular , Espermatogénesis , Espermatogonias/fisiología , Testículo/citología , Células Madre Germinales Adultas/metabolismo , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Separación Celular/métodos , Supervivencia Celular , Células Cultivadas , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Fenotipo , Espermatogonias/metabolismo , Factores de Tiempo , Transcriptoma
13.
Stem Cell Res Ther ; 8(1): 37, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28202061

RESUMEN

BACKGROUND: First trimester (FTM) and term human umbilical cord-derived perivascular cells (HUCPVCs), which are rich sources of mesenchymal stem cells (MSCs), can give rise to Sertoli cell (SC)-like as well as haploid germ cell (GC)-like cells in vitro using culture conditions that recapitulate the testicular niche. Gamete-like cells have been produced ex vivo using pluripotent stem cells as well as MSCs. However, the production of functional gametes from human stem cells has yet to be achieved. METHODS: Three independent lines of FTM and term HUCPVCs were cultured using a novel 5-week step-wise in vitro differentiation protocol recapitulating key physiological signals involved in testicular development. SC- and GC-associated phenotypical properties were assessed by real-time polymerase chain reaction (RT-PCR), quantitative PCR immunocytochemistry, flow cytometry, and fluorescence in-situ hybridization (FISH). Functional spermatogonial stem cell-like properties were assessed using a xenotranplantation assay. RESULTS: Within 3 weeks of differentiation, two morphologically distinct cell types emerged including large adherent cells and semi-attached round cells. Both early GC-associated markers (VASA, DAZL, GPR125, GFR1α) and SC-associated markers (FSHR, SOX9, AMH) were upregulated, and 5.7 ± 1.2% of these cells engrafted near the inner basal membrane in a xenograft assay. After 5 weeks in culture, 10-30% of the cells were haploid, had adopted a spermatid-like morphology, and expressed PRM1, Acrosin, and ODF2. Undifferentiated HUCPVCs secreted key factors known to regulate spermatogenesis (LIF, GDNF, BMP4, bFGF) and 10-20% of HUCPVCs co-expressed SSEA4, CD9, CD90, and CD49f. We hypothesize that the paracrine properties and cellular heterogeneity of HUCPVCs may explain their dual capacity to differentiate to both SC- and GC-like cells. CONCLUSIONS: HUCPVCs recapitulate elements of the testicular niche including their ability to differentiate into cells with Sertoli-like and haploid spermatid-like properties in vitro. Our study supports the importance of generating a niche-like environment under ex vivo conditions aiming at creating mature GC, and highlights the plasticity of HUCPVCs. This could have future applications for the treatment of some cases of male infertility.


Asunto(s)
Células Madre Mesenquimatosas/citología , Comunicación Paracrina/genética , Células de Sertoli/citología , Espermátides/citología , Espermatogénesis/genética , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Sangre Fetal/citología , Sangre Fetal/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Cultivo Primario de Células , Células de Sertoli/metabolismo , Espermátides/metabolismo
14.
Reproduction ; 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27780883

RESUMEN

The expansion of functional testicular biopsy-derived human spermatogonial stem cells (hSSC) ex-vivo may enable the restoration of fertility in pre-pubertal males having undergone gonadotoxic therapies or men with severe male factor infertility. Various somatic cells are known to regulate SSC homeostasis and spermatogenesis in the developing and adult testis. Prior attempts to recapitulate this niche demonstrated the requirement of feeder cells, such as endogenous testicular somatic cells, for germ cell expansion ex-vivo. However, this strategy has limitations for the expansion of hSSCs from tissue biopsies where spermatogenesis is absent or defective. Our aim was to evaluate first trimester human umbilical cord perivascular cells (FTM HUCPVCs), a novel source of mesenchymal stromal-like cells (MSCs), as potential human feeder cells for standardized hSSC expansion ex-vivo. Targeted RNA sequencing analysis demonstrated that CD90+ve FTM HUCPVCs expanded in vitro under germ cell culture conditions express a profile of targeted testicular-associated transcripts that is similar to cultured human CD90+ve testicular adherent cells (hTACs) and secrete LIF, FGF2 and BMP4, key growth factors known to regulate spermatogenesis. We also demonstrated that mitotically-inactivated FTM HUCPVCs support the expansion of mouse germ cells and putative SSCs ex-vivo, and that FTM HUCPVC transplantation promotes in vivo germ cell regeneration following mono-2- ethylhexyl phthalate (MEHP)-induced seminiferous tubule damage in a murine model, including a partial reconstitution of tubular cellular architecture and reestablishment of DAZL and acrosin+ve germ cell layers. Together, these data suggest that FTM HUCPVCs have phenotypical and functional properties that may support repair of the human testicular niche.

15.
Stem Cells Int ; 2016: 7513252, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27123009

RESUMEN

Myocardial infarction (MI) causes an extensive loss of heart muscle cells and leads to congestive heart disease (CAD), the leading cause of mortality and morbidity worldwide. Mesenchymal stromal cell- (MSC-) based cell therapy is a promising option to replace invasive interventions. However the optimal cell type providing significant cardiac regeneration after MI is yet to be found. The aim of our study was to investigate the cardiomyogenic differentiation potential of first trimester human umbilical cord perivascular cells (FTM HUCPVCs), a novel, young source of immunoprivileged mesenchymal stromal cells. Based on the expression of cardiomyocyte markers (cTnT, MYH6, SIRPA, and CX43) FTM and term HUCPVCs achieved significantly increased cardiomyogenic differentiation compared to bone marrow MSCs, while their immunogenicity remained significantly lower as indicated by HLA-A and HLA-G expression and susceptibility to T cell mediated cytotoxicity. When applying aggregate-based differentiation, FTM HUCPVCs showed increased aggregate formation potential and generated contracting cells within 1 week of coculture, making them the first MSC type with this ability. Our results indicate that young FTM HUCPVCs have superior cardiomyogenic potential coupled with beneficial immunogenic properties when compared to MSCs of older tissue sources, suggesting that in vitro predifferentiation could be a potential strategy to increase their effectiveness in vivo.

16.
Cell Stem Cell ; 13(5): 564-76, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24209760

RESUMEN

The mechanisms that regulate the establishment of adult stem cell pools during normal and perturbed mammalian development are still largely unknown. Here, we asked whether a maternal cytokine surge, which occurs during human maternal infections and has been implicated in cognitive disorders, might have long-lasting consequences for neural stem cell pools in adult progeny. We show that transient, maternally administered interleukin-6 (IL-6) resulted in an expanded adult forebrain neural precursor pool and perturbed olfactory neurogenesis in offspring months after fetal exposure. This increase is likely the long-term consequence of acute hyperactivation of an endogenous autocrine/paracrine IL-6-dependent self-renewal pathway that normally regulates the number of forebrain neural precursors. These studies therefore identify an IL-6-dependent neural stem cell self-renewal pathway in vivo, and support a model in which transiently increased maternal cytokines can act through this pathway in offspring to deregulate neural precursor biology from embryogenesis throughout life.


Asunto(s)
Interleucina-6/farmacología , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Femenino , Humanos , Inmunohistoquímica , Interleucina-6/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
17.
Stem Cells Dev ; 22(17): 2425-39, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23557155

RESUMEN

Human umbilical cord-derived perivascular cells (PVCs) are a recently characterized source of mesenchymal stromal cells that has gained much interest in the field of cellular therapeutics. However, very little is known about the changes in fate potential and restrictions that these cells undergo during gestational development. This study is the first to examine the phenotypic, molecular, and functional properties of first trimester (FTM)-derived PVCs, outlining properties that are unique to this population when compared to term (TERM) counterparts. FTM- and TERM-PVCs displayed analogous mesenchymal, perivascular, and immunological immunophenotypes. Both PVCs could be maintained in culture without alteration to these phenotypes or mesenchymal lineage differentiation potential. Some unique features of FTM-PVCs were uncovered in this study: (1) while the gene signatures of FTM- and TERM-PVCs were similar, key differences were observed, namely, that the Oct4A and Sox17 proteins were detected in FTM-PVCs, but not in TERM counterparts; (2) FTM-PVCs exhibited a greater proliferative potential; and (3) FTM-PVCs were more efficient in their in vitro differentiation toward selective mesenchymal cell types, including the chondrogenic and adipogenic lineages, as well as toward neuronal- and hepatocyte-like lineages, when compared to TERM-PVCs. Both PVCs were able to generate osteocytes and cardiomyocyte-like cells with similar efficiencies in vitro. Overall, FTM-PVCs show more plasticity than TERM-PVCs with regard to fate acquisition, suggesting that a restriction in multipotentiality is imposed on PVCs as gestation progresses. Taken together, our findings support the idea that PVCs from earlier in gestation may be better than later sources of multipotent stromal cells (MSCs) for some regenerative medicine applications.


Asunto(s)
Diferenciación Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana/citología , Células Madre Mesenquimatosas/citología , Pericitos/citología , Adipocitos/metabolismo , Biomarcadores/metabolismo , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Condrocitos/metabolismo , Hepatocitos/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Osteocitos/metabolismo , Factores de Transcripción SOXF/metabolismo
18.
Curr Biol ; 20(22): 2058-65, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21074438

RESUMEN

Increasing evidence suggests that deficits in adult stem cell maintenance cause aberrant tissue repair and premature aging [1]. While the mechanisms regulating stem cell longevity are largely unknown, recent studies have implicated p53 and its family member p63. Both proteins regulate organismal aging [2-4] as well as survival and self-renewal of tissue stem cells [5-9]. Intriguingly, haploinsufficiency for a third family member, p73, causes age-related neurodegeneration [10]. While this phenotype is at least partially due to loss of the ΔNp73 isoform, a potent neuronal prosurvival protein [11-16], a recent study showed that mice lacking the other p73 isoform, TAp73, have perturbations in the hippocampal dentate gyrus [17], a major neurogenic site in the adult brain. These findings, and the link between the p53 family, stem cells, and aging, suggest that TAp73 might play a previously unanticipated role in maintenance of neural stem cells. Here, we have tested this hypothesis and show that TAp73 ensures normal adult neurogenesis by promoting the long-term maintenance of neural stem cells. Moreover, we show that TAp73 does this by transcriptionally regulating the bHLH Hey2, which itself promotes neural precursor maintenance by preventing premature differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Células-Madre Neurales/metabolismo , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Senescencia Celular/genética , Senescencia Celular/fisiología , Giro Dentado/citología , Giro Dentado/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/fisiología , Regulación de la Expresión Génica , Hipocampo/metabolismo , Ratones , Datos de Secuencia Molecular , Células-Madre Neurales/citología , Neurogénesis/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Fenotipo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
19.
Dev Cell ; 18(1): 114-25, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20152182

RESUMEN

Increasing evidence indicates that epigenetic changes regulate cell genesis. Here, we ask about neural precursors, focusing on CREB binding protein (CBP), a histone acetyltransferase that, when haploinsufficient, causes Rubinstein-Taybi syndrome (RTS), a genetic disorder with cognitive dysfunction. We show that neonatal cbp(+/-) mice are behaviorally impaired, displaying perturbed vocalization behavior. cbp haploinsufficiency or genetic knockdown with siRNAs inhibited differentiation of embryonic cortical precursors into all three neural lineages, coincident with decreased CBP binding and histone acetylation at promoters of neuronal and glial genes. Inhibition of histone deacetylation rescued these deficits. Moreover, CBP phosphorylation by atypical protein kinase C zeta was necessary for histone acetylation at neural gene promoters and appropriate differentiation. These data support a model in which environmental cues regulate CBP activity and histone acetylation to control neural precursor competency to differentiate, and indicate that cbp haploinsufficiency disrupts this mechanism, thereby likely causing cognitive dysfunction in RTS.


Asunto(s)
Encéfalo/anomalías , Encéfalo/enzimología , Proteína de Unión a CREB/metabolismo , Malformaciones del Sistema Nervioso/enzimología , Neurogénesis/fisiología , Síndrome de Rubinstein-Taybi/enzimología , Acetilación , Animales , Encéfalo/fisiopatología , Proteína de Unión a CREB/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Histonas/genética , Histonas/metabolismo , Trastornos Mentales/enzimología , Trastornos Mentales/genética , Trastornos Mentales/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/fisiopatología , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Interferencia de ARN , Síndrome de Rubinstein-Taybi/genética , Síndrome de Rubinstein-Taybi/fisiopatología , Células Madre/enzimología
20.
Cell Stem Cell ; 4(6): 507-10, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19497279

RESUMEN

Adult neural stem cells (NSCs) are involved in regulating mammalian behavior and are controlled by diverse external stimuli. Improved understanding of the physical location of NSCs and the microenvironmental cues that regulate their behavior, which combine to define the NSC "home," or niche, may reveal how to control their function.


Asunto(s)
Neuronas/citología , Nicho de Células Madre/fisiología , Células Madre Adultas/fisiología , Animales , Conducta/fisiología , Humanos , Nicho de Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA