Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Cell Tissue Res ; 389(2): 289-308, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35624315

RESUMO

Stem cells are known to have excellent regenerative ability, which is primarily facilitated by indirect paracrine factors, rather than via direct cell replacement. The regenerative process is mediated by the release of extracellular matrix molecules, cytokines, and growth factors, which are also present in the media during cultivation. Herein, we aimed to demonstrate the functionality of key factors and mechanisms in skin regeneration through the analysis of conditioned media derived from fetal stem cells. A series of processes, including 3D pellet cultures, filtration and lyophilization is developed to fabricate human fetal cartilage-derived progenitor cells-conditioned media (hFCPCs-CM) and its useful properties are compared with those of human bone marrow-derived MSCs-conditioned media (hBMSCs-CM) in terms of biochemical characterization, and in vitro studies of fibroblast behavior, macrophage polarization, and burn wound healing. The hFCPCs-CM show to be devoid of cellular components but to contain large amounts of total protein, collagen, glycosaminoglycans, and growth factors, including IGFBP-2, IGFBP-6, HGF, VEGF, TGF ß3, and M-CSF, and contain a specific protein, collagen alpha-1(XIV) compare with hBMSCs-CM. The therapeutic potential of hFCPCs-CM observes to be better than that of hBMSCs-CM in the viability, proliferation, and migration of fibroblasts, and M2 macrophage polarization in vitro, and efficient acceleration of wound healing and minimization of scar formation in third-degree burn wounds in a rat model. The current study shows the potential therapeutic effect of hFCPCs and provides a rationale for using the secretome released from fetal progenitor cells to promote the regeneration of skin tissues, both quantitatively and qualitatively. The ready-to-use product of human fetal cartilage-derived progenitor cells-conditioned media (hFCPCs-CM) are fabricated via a series of techniques, including a 3D culture of hFCPCs, filtration using a 3.5 kDa cutoff dialysis membrane, and lyophilization of the CM. hFCPCs-CM contains many ECM molecules and biomolecules that improves wound healing through efficient acceleration of M2 macrophage polarization and reduction of scar formation.


Assuntos
Queimaduras , Células-Tronco Fetais , Animais , Queimaduras/patologia , Queimaduras/terapia , Cicatriz/patologia , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Meios de Cultivo Condicionados/farmacologia , Células-Tronco Fetais/metabolismo , Fibroblastos/metabolismo , Humanos , Ratos , Pele/patologia , Células-Tronco , Cicatrização
2.
Int J Mol Sci ; 22(7)2021 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-33918297

RESUMO

We previously reported that c-KIT+ human amniotic-fluid derived stem cells obtained from leftover samples of routine II trimester prenatal diagnosis (fetal hAFS) are endowed with regenerative paracrine potential driving pro-survival, anti-fibrotic and proliferative effects. hAFS may also be isolated from III trimester clinical waste samples during scheduled C-sections (perinatal hAFS), thus offering a more easily accessible alternative when compared to fetal hAFS. Nonetheless, little is known about the paracrine profile of perinatal hAFS. Here we provide a detailed characterization of the hAFS total secretome (i.e., the entirety of soluble paracrine factors released by cells in the conditioned medium, hAFS-CM) and the extracellular vesicles (hAFS-EVs) within it, from II trimester fetal- versus III trimester perinatal cells. Fetal- and perinatal hAFS were characterized and subject to hypoxic preconditioning to enhance their paracrine potential. hAFS-CM and hAFS-EV formulations were analyzed for protein and chemokine/cytokine content, and the EV cargo was further investigated by RNA sequencing. The phenotype of fetal- and perinatal hAFS, along with their corresponding secretome formulations, overlapped; yet, fetal hAFS showed immature oxidative phosphorylation activity when compared to perinatal ones. The profiling of their paracrine cargo revealed some differences according to gestational stage and hypoxic preconditioning. Both cell sources provided formulations enriched with neurotrophic, immunomodulatory, anti-fibrotic and endothelial stimulating factors, and the immature fetal hAFS secretome was defined by a more pronounced pro-vasculogenic, regenerative, pro-resolving and anti-aging profile. Small RNA profiling showed microRNA enrichment in both fetal- and perinatal hAFS-EV cargo, with a stably- expressed pro-resolving core as a reference molecular signature. Here we confirm that hAFS represents an appealing source of regenerative paracrine factors; the selection of either fetal or perinatal hAFS secretome formulations for future paracrine therapy should be evaluated considering the specific clinical scenario.


Assuntos
Células-Tronco Fetais/metabolismo , Segundo Trimestre da Gravidez/metabolismo , Terceiro Trimestre da Gravidez/metabolismo , Proteoma , Adulto , Líquido Amniótico/citologia , Secreções Corporais , Vesículas Extracelulares/ultraestrutura , Feminino , Humanos , Hipóxia/metabolismo , Gravidez
3.
Neurotoxicol Teratol ; 83: 106943, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33221301

RESUMO

Prenatal ethanol exposure can result in loss of neural stem cells (NSCs) and decreased brain growth. Here, we assessed whether a noncoding RNA (ncRNA) related to the NSC self-renewal factor Oct4/Pou5f1, and transcribed from a processed pseudogene locus on mouse chromosome 9 (mOct4pg9), contributed to the loss of NSCs due to ethanol. Mouse fetal cortical-derived NSCs, cultured ex vivo to mimic the early neurogenic environment of the fetal telencephalon, expressed mOct4pg9 ncRNA at significantly higher levels than the parent Oct4/Pou5f1 mRNA. Ethanol exposure increased expression of mOct4pg9 ncRNA, but decreased expression of Oct4/Pou5f1. Gain- and loss-of-function analyses indicated that mOct4pg9 overexpression generally mimicked effects of ethanol exposure, resulting in increased proliferation and expression of transcripts associated with neural maturation. Moreover, mOct4pg9 associated with Ago2 and with miRNAs, including the anti-proliferative miR-328-3p, whose levels were reduced following mOct4pg9 overexpression. Finally, mOct4pg9 inhibited Oct4/Pou5f1-3'UTR-dependent protein translation. Consistent with these observations, data from single-cell transcriptome analysis showed that mOct4pg9-expressing progenitors lack Oct4/Pou5f1, but instead overexpress transcripts for increased mitosis, suggesting initiation of transit amplification. Collectively, these data suggest that the inhibitory effects of ethanol on brain development are explained, in part, by a novel ncRNA which promotes loss of NSC identity and maturation.


Assuntos
Etanol/toxicidade , Células-Tronco Fetais/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Fator 3 de Transcrição de Octâmero/genética , RNA não Traduzido/genética , Animais , Proteínas Argonautas/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Transtornos do Espectro Alcoólico Fetal/genética , Transtornos do Espectro Alcoólico Fetal/metabolismo , Transtornos do Espectro Alcoólico Fetal/patologia , Células-Tronco Fetais/metabolismo , Células-Tronco Fetais/patologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Fator 3 de Transcrição de Octâmero/antagonistas & inibidores , Fator 3 de Transcrição de Octâmero/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Pseudogenes , RNA não Traduzido/metabolismo , Análise de Célula Única
4.
Res Vet Sci ; 133: 239-245, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33032111

RESUMO

The liver plays essential roles in human and animal organisms, such as the storage, release, metabolism, and elimination of various endogenous or exogenous substances. Although its vital importance, few treatments are yet available when a hepatic failure occurs, and hence, the use of stem cells has arisen as a possible solution for both human and veterinary medicines. Previous studies have shown the existence of hepatic progenitor cells in human fetuses that were positive for EpCAM and NCAM. There is limited evidence, however, further identification and characterization of these cells in other species. Considering the similarity between dogs and humans regarding physiology, and also the increasing importance of developing new treatments for both veterinary and translational medicine, this study attempted to identify hepatic progenitor cells in canine fetal liver. For that, livers from canine fetuses were collected, cells were isolated by enzymatic digestion and cultured. Cells were characterized regarding morphology and expression of EpCAM, NCAM, Nestin, and Thy-1/CD90 markers. Our results suggest that it is possible to identify hepatic progenitor cells in the canine fetal liver; however, for therapeutic use, further techniques for cellular isolation and culture are necessary to obtain enriched populations of hepatic progenitors from the canine fetal liver.


Assuntos
Cães/embriologia , Células-Tronco Fetais/citologia , Fígado/embriologia , Animais , Biomarcadores/metabolismo , Células Cultivadas , Cães/anatomia & histologia , Molécula de Adesão da Célula Epitelial/metabolismo , Células-Tronco Fetais/metabolismo , Feto/citologia , Feto/embriologia , Hepatócitos/metabolismo , Fígado/citologia , Fígado/metabolismo , Antígenos Thy-1/metabolismo
5.
Stem Cell Reports ; 15(4): 869-882, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32976765

RESUMO

Human glial progenitor cells (hGPCs) are promising cellular substrates to explore for the in situ production of new neurons for brain repair. Proof of concept for direct neuronal reprogramming of glial progenitors has been obtained in mouse models in vivo, but conversion using human cells has not yet been demonstrated. Such studies have been difficult to perform since hGPCs are born late during human fetal development, with limited accessibility for in vitro culture. In this study, we show proof of concept of hGPC conversion using fetal cells and also establish a renewable and reproducible stem cell-based hGPC system for direct neural conversion in vitro. Using this system, we have identified optimal combinations of fate determinants for the efficient dopaminergic (DA) conversion of hGPCs, thereby yielding a therapeutically relevant cell type that selectively degenerates in Parkinson's disease. The induced DA neurons show a progressive, subtype-specific phenotypic maturation and acquire functional electrophysiological properties indicative of DA phenotype.


Assuntos
Reprogramação Celular , Neurônios Dopaminérgicos/citologia , Células-Tronco Fetais/citologia , Mesencéfalo/citologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios Dopaminérgicos/metabolismo , Células-Tronco Fetais/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
6.
Regen Med ; 15(6): 1719-1733, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32772793

RESUMO

Aim: To investigate direct roles of TGF-ß1 signaling in the differentiation process of fetal hepatic progenitor cells (HPCs). Materials & methods: Exogenous TGF-ß1 and SB431542 were added into fetal HPCs. Then, SB431542 was intraperitoneally injected into pregnant mice for 8 days. Results: Fetal HPCs treated with TGF-ß1 differentiated into cholangiocytes. However, hepatocyte marker was highly expressed after inhibiting TGF-ß1 signaling. In vivo, hematopoietic cells were gradually replaced with liver cells and TGF-ß1 expression was evidently decreased as fetal liver developed. Inhibition of TGF-ß1 signaling caused increase of ALB+ cells, but CK19 expression was more obvious in control mice livers. Conclusion: TGF-ß1 signaling may play decisive roles in fetal HPCs differentiation into functional hepatocytes or cholangiocytes.


Assuntos
Diferenciação Celular , Células-Tronco Fetais/citologia , Hepatócitos/citologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Feminino , Células-Tronco Fetais/metabolismo , Hepatócitos/metabolismo , Camundongos , Transdução de Sinais
7.
Methods Mol Biol ; 2171: 231-236, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32705645

RESUMO

Intestinal organoids are useful models for studying the characteristics of intestinal diseases and their treatment. However, a major limiting factor in their usability is the need for donor tissue fragments or pluripotent stem cells to generate the organoids. Here, we describe an approach to generate intestinal organoids from fibroblasts, a new source. We used direct reprogramming technology to generate cells with the properties of fetal intestine-derived progenitor cells (FIPCs) from mouse embryonic fibroblasts (MEFs). These induced FIPCs (iFIPCs) can give rise to cells resembling intestinal stem cells (ISCs), henceforth referred to as induced ISCs (iISCs). These iFIPCs and iISCs form spherical and budding organoids, respectively, similar to FIPCs and ISCs. These induced intestinal organoids could be used for studies on intestinal diseases and regenerative therapy.


Assuntos
Células-Tronco Fetais/citologia , Fibroblastos/citologia , Mucosa Intestinal/citologia , Organoides/citologia , Células-Tronco/citologia , Animais , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Células-Tronco Fetais/metabolismo , Fibroblastos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Organoides/metabolismo , Células-Tronco/metabolismo
8.
Mol Ther ; 28(7): 1645-1657, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32353323

RESUMO

Retinal pigment epithelial (RPE) cell replacement therapy has provided promising outcomes in the treatment of retinal degenerative diseases (RDDs), but the resulting limited visual improvement has raised questions about graft survival and differentiation. Through combined treatment with vitamin C and valproic acid (together, VV), we activated human fetal RPE (fRPE) cells to become highly proliferative fetal RPE stem-like cells (fRPESCs). In this study, we report that SOX2 (SRY-box 2) activation contributed to mesenchymal-epithelial transition and elevated the retinal progenitor and mesenchymal stromal markers expressions of fRPESCs. These fRPESCs could differentiate into RPE cells, rod photoreceptors, and mesenchymal lineage progenies under defined conditions. Finally, fRPESCs were transplanted into the subretinal space of an RDD mouse model, and a photoreceptor rescue benefit was demonstrated. The RPE and rod photoreceptor differentiation of transplanted fRPESCs may account for the neural retinal recovery. This study establishes fRPESCs as a highly proliferative, multi-lineage differentiation potential (including RPE, rod photoreceptor, and mesenchymal lineage differentiation), mesenchymal-to-epithelial-transitioned retinal stem-like cell source for cell-based therapy of RDDs.


Assuntos
Ácido Ascórbico/farmacologia , Células-Tronco Fetais/transplante , Degeneração Retiniana/terapia , Epitélio Pigmentado da Retina/embriologia , Fatores de Transcrição SOXB1/metabolismo , Ácido Valproico/farmacologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Células-Tronco Fetais/citologia , Células-Tronco Fetais/efeitos dos fármacos , Células-Tronco Fetais/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Degeneração Retiniana/metabolismo , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Resultado do Tratamento , Regulação para Cima
9.
Stem Cell Rev Rep ; 16(3): 524-540, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32020407

RESUMO

Nongonadal tissues express luteinizing hormone-chorionic gonadotropin receptors (LHCG-R) which are essential for their growth during fetal development. Adult mesenchymal stem/stromal cells (MSCs) have been shown to express functional LHCG-R outside pregnancy conditions, making them susceptible to hCG stimulation. In the present study we tested the effect of hCG treatment on bone marrow (BM) derived adherent stem cells in vitro, isolated from a parous women, mother of male sons, in order to evaluate its effect on maternal MSCs and in the same time on fetal microchimeric stem cells (FMSCs), to better understand the outcomes of this safe and affordable treatment on cell proliferation and expression of pluripotency genes. Our study highlights the beneficial effects of hCG exposure on gene regulation in bone marrow adherent stem cells through the upregulation of pluripotency genes and selection of more primitive mesenchymal stem cells with a better differentiation potential. Validation of these effects on MSCs and FMSCs long after parturition in vivo represents a close perspective as it could set the premises of a new mobilization strategy for the stem cell transplantation procedures in the clinical setting.


Assuntos
Células da Medula Óssea/citologia , Quimerismo/efeitos dos fármacos , Gonadotropina Coriônica/farmacologia , Células-Tronco Fetais/citologia , Células-Tronco Fetais/imunologia , Tolerância Imunológica/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Separação Celular , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Feminino , Células-Tronco Fetais/efeitos dos fármacos , Células-Tronco Fetais/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Osteogênese/efeitos dos fármacos , Osteogênese/genética
10.
Stem Cell Res Ther ; 10(1): 383, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31843019

RESUMO

BACKGROUND: Radiation dermatitis is a refractory skin injury caused by radiotherapy. Human fetal skin-derived stem cell (hFSSC) is a preferable source for cell therapy and skin tissue regeneration. In the present study, we investigated the repair effect of using hFSSC secretome on a radiation skin injury model in rats. METHODS: We prepared the hFSSC secretome and studied its effects on the proliferation and tube formation of human umbilical vein endothelial cell (HUVEC) in vitro. Furthermore, we used a Sr-90 radiation-induced skin injury model of rats and evaluated the effects of hFSSC secretome on radiation skin injury in vivo. RESULTS: The results showed that hFSSC secretome significantly promoted the proliferation and tube formation of HUVEC in vitro; in addition, hFSSC secretome-treated rats exhibited higher healing quality and faster healing rate than the other two control groups; the expression level of collagen type III α 1 (Col3A1), transforming growth factor ß3 (TGF-ß3), angiotensin 1 (Ang-1), angiotensin 2 (Ang-2), vascular endothelial growth factor (VEGF), and placental growth factor (PLGF) was significantly increased, while collagen type I α 2 (Col1A2) and transforming growth factor ß1 (TGF-ß1) were decreased in hFSSC secretome group. CONCLUSIONS: In conclusion, our results provided the first evidence on the effects of hFSSC secretome towards radiation-induced skin injury. We found that hFSSC secretome significantly enhanced radiation dermatitis angiogenesis, and the therapeutic effects could match with the characteristics of fetal skin. It may act as a kind of novel cell-free therapeutic approach for radiation-induced cutaneous wound healing.


Assuntos
Células-Tronco Fetais/metabolismo , Radioterapia/efeitos adversos , Dermatopatias/induzido quimicamente , Animais , Proliferação de Células , Modelos Animais de Doenças , Humanos , Camundongos , Neovascularização Fisiológica
11.
Sci Rep ; 9(1): 9973, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31292491

RESUMO

EPNs comprise a heterogeneous group of neuroepithelial tumors, accounting for about 10% of all intracranial tumors in children and up to 30% of brain tumors in those younger than 3 years. Actually, the pattern therapy for low-grade EPNs includes complete surgical resection followed by radiation therapy. Total surgical excision is often not possible due to tumor location. The aim of this study was to evaluate, for the first time, the anti-tumor activity of Amblyomin-X in 4 primary cultures derived from pediatric anaplastic posterior fossa EPN, Group A (anaplastic, WHO grade III) and one primary culture of a high grade neuroepithelial tumor with MN1 alteration, which was initially misdiagnosed as EPN: i) by in vitro assays: comparisons of temozolomide and cisplatin; ii) by intracranial xenograft model. Amblyomin-X was able to induce cell death in EPN cells in a more significant percentage compared to cisplatin. The cytotoxic effects of Amblyomin-X were not detected on hFSCs used as control, as opposed to cisplatin-treatment, which promoted a substantial effect in the hAFSCs viability. TEM analysis showed ultrastructural alterations related to the process of cell death: mitochondrial degeneration, autophagosomes and aggregate-like structures. MRI and histopathological analyzes demonstrated significant tumor mass regression. Our results suggest that Amblyomin-X has a selective effect on tumor cells by inducing apoptotic cell death and may be a therapeutic option for Group AEPNs.


Assuntos
Antineoplásicos/farmacologia , Ependimoma/tratamento farmacológico , Proteínas e Peptídeos Salivares/farmacologia , Adulto , Animais , Apoptose/efeitos dos fármacos , Proteínas de Artrópodes , Criança , Pré-Escolar , Feminino , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Humanos , Masculino , Ratos Wistar , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
12.
Int J Dev Biol ; 63(6-7): 295-299, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31250913

RESUMO

We have successfully isolated cells with stem-like properties from bottlenose dolphin (Tursiops truncatus) umbilical cord. Our results show that this cetacean species has embryonic fetal and adult stem cells as do humans and other studied mammals. This accomplishment allows to eventually investigate whether dolphins, due to their unique adaptations to aquatic environments, have special stem cell lineages or distinctive mechanisms of cell programming. Further characterization of their potency to differentiate into multiple cell lineages would fulfill numerous applicative purposes. We characterized, developed and refined a new protocol for obtaining potential stem cells from umbilical cord tissues of the bottlenose dolphin. Tissue samples were taken from umbilical cords of successful deliveries immediately after placenta ejection and collection from the water. Umbilical cord samples (2-3 cm3) were excised and subjected to enzymatic digestion and mechanical dissociation. Viable cells from specimens resident in the Oceanografic Valencia were cultured and subsequently isolated and tested for pluripotent characteristics (cell morphology, phenotype and expression of surface markers). Cell viability was confirmed also after freezing/thawing. The established protocol is suitable for collection/isolation/culture of dolphin potential mesenchymal stem cells from dolphin umbilical cord, which can be deposited in cell banks for future research needs.


Assuntos
Células-Tronco Adultas/citologia , Golfinho Nariz-de-Garrafa/metabolismo , Separação Celular/métodos , Células-Tronco Embrionárias/citologia , Células-Tronco Fetais/citologia , Células-Tronco Mesenquimais/citologia , Cordão Umbilical/citologia , Células-Tronco Adultas/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Feminino , Células-Tronco Fetais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Cordão Umbilical/metabolismo
13.
Am J Physiol Cell Physiol ; 317(2): C348-C357, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31166709

RESUMO

Maternal endothelial dysfunction is a cental feature of preeclampsia (PE), a hypertensive disorder of pregnancy. Factors in the maternal circulation are thought to contribute to this endothelial dysfunction. Although understudied, factors in the fetal circulation may influence fetal endothelial cell interactions with endothelial progenitor cells as critical steps in placental angiogenesis. We hypothesize that cell-cell interactions that are important for pregnancy health are impaired by fetal serum from PE pregnancies and that 1,25(OH)2-vitamin D3 attenuates the negative effects of this serum on cell function. We tested the ability of fetal cord blood-derived endothelial progenitor cells [endothelial colony-forming cells (ECFCs)] to invade into established monolayers and capillary tubule-like structures of human fetal umbilical venous endothelial cells (HUVECs), while in the presence/absence of fetal cord serum from uncomplicated or PE pregnancies, and tested the ability of 1,25(OH)2-vitamin D3 to modulate the serum-mediated effects. PE cord serum reduced the invasion of fetal ECFCs into HUVEC monolayers or tubule networks. Vitamin D attenuated these effects of PE fetal serum on endothelial functional properties. Immunocytochemical studies revealed involvement of VE-cadherin contacts in interactions between ECFCs and mature fetal endothelial cells. PE cord serum reduces the ability of fetal endothelial progenitor cells to incorporate into fetal endothelial cell networks. Physiologic concentrations of vitamin D reverse these PE serum-mediated effects. These data appear consistent with lines of evidence that vitamin D has antipreeclampsia effects.


Assuntos
Calcitriol/farmacologia , Comunicação Celular/efeitos dos fármacos , Células Progenitoras Endoteliais/efeitos dos fármacos , Células-Tronco Fetais/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Pré-Eclâmpsia/tratamento farmacológico , Adulto , Estudos de Casos e Controles , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/metabolismo , Células Progenitoras Endoteliais/metabolismo , Feminino , Células-Tronco Fetais/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Pré-Eclâmpsia/metabolismo , Gravidez , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Transdução de Sinais
14.
Stem Cell Rev Rep ; 15(4): 519-529, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31123983

RESUMO

Fetal-maternal microchimerism describes the acquisition of fetal stem cells (FSC) by the mother during pregnancy and their long-term persistence after parturition. FSC may engraft in a variety of maternal tissues especially if there is organ/tissue injury, but their role and mechanism of persistence still remains elusive. Clinical applications due to their pluripotency, immunomodulatory effects and accessibility make them good candidates for ex-vivo manipulation and autologous therapies. The hair follicles contain a distinctive niche for pluripotent stem cells (PSC). To date, there is no published evidence of fetal microchimerism in the hair follicle. In our study, follicular unit extraction (FUE) technique allowed easy stem cell cultures to be obtained while simple hair follicle removal by pull-out technique failed to generate stem cells in culture. We identified microchimeric fetal stem cells within the primitive population of maternal stem cells isolated from the hair follicles with typical mesenchymal phenotype, expression of PSC genes and differentiation potential towards osteocytes, adypocites and chondrocytes. This is the first study to isolate fetal microchimeric stem cells in adult human hair long after parturition. We presume a sanctuary partition mechanism with PSC of the mother deposited during early embryogenesis could explain their long-term persistence.


Assuntos
Diferenciação Celular , Quimerismo , Células-Tronco Fetais , Folículo Piloso , Adulto , Feminino , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Feto/citologia , Feto/metabolismo , Folículo Piloso/citologia , Folículo Piloso/metabolismo , Humanos , Gravidez
15.
Curr Top Dev Biol ; 132: 1-29, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30797507

RESUMO

Hematopoiesis is the process by which mature blood and immune cells are produced from hematopoietic stem and progenitor cells (HSCs and HSPCs). The last several decades of research have shed light on the origin of HSCs, as well as the heterogeneous pools of fetal progenitors that contribute to lifelong hematopoiesis. The overarching concept that hematopoiesis occurs in dynamic, overlapping waves throughout development, with each wave contributing to both continuous and developmentally limited cell types, has been solidified over the years. However, recent advances in our ability to track the production of hematopoietic cells in vivo have challenged several long-held dogmas on the origin and persistence of distinct hematopoietic cell types. In this review, we highlight emerging concepts in hematopoietic development and identify unanswered questions.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Animais , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Fetais/metabolismo , Humanos
16.
J Pathol ; 248(2): 155-163, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30680725

RESUMO

The cellular mechanisms by which hepatitis C virus (HCV) replication might mediate cytopathic effects are controversial and not entirely clear. In this study, we found that blood-borne HCV (bbHCV) infection could lead to endoplasmic reticulum (ER)-stress and mitochondria-related/caspase-dependent apoptosis at the early stages of infection based on use of the highly efficient bbHCV cell culture model established previously. Sections of bbHCV-infected human fetal liver stem cells (hFLSCs) revealed convolution and nonlinear ER, cell vacuolization, swelling of mitochondria, and numerous double membrane vesicles (DMVs). The percentage of apoptotic hFLSCs infected by bbHCV reached 29.8% at 16 h postinfection, and the amount of cytochrome c increased remarkably in the cytosolic protein fraction. However, over time, apoptosis was inhibited due to the activation of NF-κB. The expression of NF-κB-p65, Bcl-xL, XIAP, and c-FLIPL in hFLSCs was increased significantly 24 h after in infection by bbHCV. The accelerated cell death cycles involving apoptosis, regeneration and repair by bbHCV infection might give rise to the development of cirrhosis, and ultimately to hepatocellular carcinogenesis. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Células-Tronco Fetais/patologia , Hepacivirus/crescimento & desenvolvimento , Hepatite C Crônica/virologia , Fígado/patologia , Replicação Viral , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Células-Tronco Fetais/metabolismo , Células-Tronco Fetais/virologia , Hepatite C Crônica/metabolismo , Hepatite C Crônica/patologia , Humanos , Fígado/metabolismo , Fígado/virologia , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/patologia , Mitocôndrias Hepáticas/virologia , Estresse Oxidativo , Transdução de Sinais
17.
Sci Rep ; 8(1): 9321, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29915318

RESUMO

Human multipotent mesenchymal stromal cells (MSC) are isolated from a plethora of tissue sources for cell therapy purposes. In 2006, the International Society for Cellular Therapy (ISCT) published minimal guidelines to define MSC identity. Nevertheless, many independent studies demonstrated that cells meeting the ISCT criteria possessed heterogeneous phenotypes and functionalities, heavily influenced by culture conditions. In this study, human MSC derived from many adult (bone marrow and adipose tissue) or fetal (cord blood, Wharton's jelly, umbilical cord perivascular compartment and amniotic fluid) tissues were investigated. Their immunophenotype was analyzed to define consistent source-specific markers by extensive flow cytometry analysis and real-time qRT-PCR. CD271+ subpopulations were detected in adult MSC, whereas NG2 was significantly more expressed in fetal MSC but failed validation on independent samples coming from an external laboratory. The highest number of CD271+ adult MSC were detected soon after isolation in serum-based culture conditions. Furthermore, heterogeneous percentages of CD271 expression were found in platelet lysate-based or serum-free culture conditions. Finally, CD271+ adult MSC showed high clonogenic and osteogenic properties as compared to CD271- cells. To conclude, in this phenotype-function correlation study CD271+ subpopulation confers heterogeneity on adult MSC, confirming the need of more specific markers to address MSC properties.


Assuntos
Adapaleno/metabolismo , Células-Tronco Adultas/metabolismo , Células-Tronco Fetais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Adulto , Biomarcadores/metabolismo , Forma Celular , Células Cultivadas , Células Clonais , Análise por Conglomerados , Humanos , Imunofenotipagem , Pessoa de Meia-Idade , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo
18.
Sci China Life Sci ; 61(8): 885-892, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29934917

RESUMO

Fetal liver (FL) is an intricate and highly vascularized hematopoietic organ, which can support the extensive expansion of hematopoietic stem cells (HSCs) without loss of stemness, as well as of the downstream lineages of HSCs. This powerful function of FL largely benefits from the niche (or microenvironment), which provides a residence for HSC expansion. Numerous studies have demonstrated that the FL niche consists of heterogeneous cell populations that associate with HSCs spatially and regulate HSCs functionally. At the molecular level, a complex of cell extrinsic and intrinsic signaling network within the FL niche cells maintains HSC expansion. Here, we summarize recent studies on the analysis of the FL HSCs and their niche, and specifically on the molecular regulatory network for HSC expansion. Based on these studies, we hypothesize a strategy to obtain a large number of functional HSCs via 3D reconstruction of FL organoid ex vivo for clinical treatment in the future.


Assuntos
Proliferação de Células , Células-Tronco Fetais/citologia , Células-Tronco Hematopoéticas/citologia , Fígado/citologia , Animais , Células-Tronco Fetais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fígado/irrigação sanguínea , Fígado/embriologia , Nicho de Células-Tronco/genética
19.
Stem Cells ; 36(10): 1475-1486, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29893019

RESUMO

This study reviewed the occurrence of chondroitin sulfate (CS) motifs 4-C-3, 7-D-4, and 3-B-3(-), which are expressed by progenitor cells in tissues undergoing morphogenesis. These motifs have a transient early expression pattern during tissue development and also appear in mature tissues during pathological remodeling and attempted repair processes by activated adult stem cells. The CS motifs are information and recognition modules, which may regulate cellular behavior and delineate stem cell niches in developmental tissues. One of the difficulties in determining the precise role of stem cells in tissue development and repair processes is their short engraftment period and the lack of specific markers, which differentiate the activated stem cell lineages from the resident cells. The CS sulfation motifs 7-D-4, 4-C-3, and 3-B-3 (-) decorate cell surface proteoglycans on activated stem/progenitor cells and appear to identify these cells in transitional areas of tissue development and in tissue repair and may be applicable to determining a more precise role for stem cells in tissue morphogenesis. Stem Cells 2018;36:1475-1486.


Assuntos
Sulfatos de Condroitina/metabolismo , Células-Tronco Fetais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteoglicanas/metabolismo , Células-Tronco/metabolismo , Diferenciação Celular , Feminino , Humanos , Masculino
20.
BMC Res Notes ; 11(1): 343, 2018 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-29843819

RESUMO

OBJECTIVE: Human amniotic epithelial cells (hAECs) are a novel source of stem cells and have immunomodulatory effects on both the innate and adoptive immune system. hAECs can differentiate into multiple cell lineages that make them a suitable cell source for regenerative medicine. These cells express multiple toll-like receptors (TLRs) and respond to various TLR ligands. This study aimed to evaluate the effect of lipopolysaccharide (LPS), a TLR4 ligand, on the level of immunomodulatory and immunostimulatory factors of hAECs. RESULTS: Our results indicated that LPS had the ability to up-regulate the expression of prostaglandin E2 synthase and transforming growth factor-beta1 in hAECs. However, there was no change in the level of interleukin-1beta, interleukin-6 and interleukin-10 in hAECs when were stimulated with LPS. In addition, we observed tumor necrosis factor-alpha was only expressed at very low level in some of hAECs samples which its expression was independent of the effects of LPS.


Assuntos
Âmnio/citologia , Citocinas/efeitos dos fármacos , Células Epiteliais , Células-Tronco Fetais , Fatores Imunológicos/farmacologia , Lipopolissacarídeos/farmacologia , Prostaglandina-E Sintases/efeitos dos fármacos , Adulto , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Células-Tronco Fetais/efeitos dos fármacos , Células-Tronco Fetais/imunologia , Células-Tronco Fetais/metabolismo , Humanos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...