Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Aging Neurosci ; 14: 925728, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35966788

RESUMEN

Background: Developing brain is highly plastic and can be easily affected. Growing pediatric usage of anesthetics during painless procedures has raised concerns about the effect of low-dose anesthetics on neurodevelopment. It is urgent to ascertain the neuronal effect of low-dose Propofol, a widely used anesthetic in pediatrics, on developing brains. Methods: The behavioral tests after neonatal exposure to low-dose/high-dose Propofol in mice were conducted to clarify the cognitive effect. The nascent cells undergoing proliferation and differentiation stage in the hippocampus and cultured neural stem cells (NSCs) were further identified. In addition, single-nuclei RNA sequencing (snRNA-seq), NSCs bulk RNA-seq, and metabolism trials were performed for pathway investigation. Furthermore, small interfering RNA and stereotactic adenovirus injection were, respectively, used in NSCs and hippocampal to confirm the underlying mechanism. Results: Behavioral tests in mice showed enhanced spatial cognitive ability after being exposed to low-dose Propofol. Activated neurogenesis was observed both in hippocampal and cultured NSCs. Moreover, transcriptome analysis of snRNA-seq, bulk RNA-seq, and metabolism trials revealed a significantly enhanced oxidative phosphorylation (OXPHOS) level in NSCs. Furthermore, PGC-1α, a master regulator in mitochondria metabolism, was found upregulated after Propofol exposure both in vivo and in vitro. Importantly, downregulation of PGC-1α remarkably prevented the effects of low-dose Propofol in activating OXPHOS and neurogenesis. Conclusions: Taken together, this study demonstrates a novel alteration of mitochondrial function in hippocampal neurogenesis after low-dose Propofol exposure, suggesting the safety, even potentially beneficial effect, of low-dose Propofol in pediatric use.

2.
Stem Cells Transl Med ; 11(6): 659-673, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35648087

RESUMEN

Interstitial cystitis (IC) is a bladder syndrome of unclear etiology with no generally accepted treatment. Growing evidence suggest that periostin (POSTN) is an important homeostatic component in the tissue repair and regeneration in adulthood, but its function in urinary bladder regeneration is still unknown. Here we investigate whether POSTN is involved in bladder tissue repair in a cyclophosphamide (CYP)-induced interstitial cystitis model. POSTN is primarily expressed in bladder stroma (detrusor smooth muscle and lamina propria) and upregulated in response to CYP-induced injury. POSTN deficiency resulted in more severe hematuria, aggravated edema of the bladder, and delayed umbrella cell recovery. Besides, less proliferative urothelial cells (labeled by pHH3, Ki67, and EdU) and lower expression of Krt14 (a urothelial stem cell marker) were detected in POSTN-/- mice post CYP exposure, indicating a limited urothelial regeneration. Further investigations revealed that POSTN could induce Wnt4 upregulation and activate AKT signaling, which together activates ß-catenin signaling to drive urothelial stem cell proliferation. In addition, POSTN can promote resident macrophage proliferation and polarization to a pro-regenerative (M2) phenotype, which favors urothelial regeneration. Furthermore, we generated injectable P-GelMA granular hydrogel as a biomaterial carrier to deliver recombinant POSTN into the bladder, which could increase urothelial stem cells number, decrease umbrella cells exfoliation, and hence alleviate hematuria in a CYP-induced interstitial cystitis model. In summary, our findings identify a pivotal role of POSTN in bladder urothelial regeneration and suggest that intravesical biomaterials-assisted POSTN delivery may be an efficacious treatment for interstitial cystitis.


Asunto(s)
Cistitis Intersticial , Cistitis , Animales , Proliferación Celular , Ciclofosfamida/efectos adversos , Ciclofosfamida/metabolismo , Cistitis/inducido químicamente , Cistitis/genética , Cistitis/metabolismo , Cistitis Intersticial/metabolismo , Hematuria/metabolismo , Macrófagos/metabolismo , Ratones , Vejiga Urinaria
3.
Front Cell Dev Biol ; 9: 642533, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33968928

RESUMEN

Objective: Many tissues contained resident mesenchymal stromal/stem cells (MSCs) that facilitated tissue hemostasis and repair. However, there is no typical marker to identify the resident cardiac MSCs. We aimed to determine if CD51 could be an optimal marker of cardiac MSCs and assess their therapeutic potential for mice with acute myocardial infarction (AMI). Methods: Cardiac-derived CD51+CD31-CD45-Ter119- cells (named CD51+cMSCs) were isolated from C57BL/6 mice(7-day-old) by flow cytometry. The CD51+cMSCs were characterized by proliferation capacity, multi-differentiation potential, and expression of typical MSC-related markers. Adult C57BL/6 mice (12-week-old) were utilized for an AMI model via permanently ligating the left anterior descending coronary artery. The therapeutic efficacy of CD51+cMSCs was estimated by echocardiography and pathological staining. To determine the underlying mechanism, lentiviruses were utilized to knock down gene (stem cell factor [SCF]) expression of CD51+cMSCs. Results: In this study, CD51 was expressed in the entire layers of the cardiac wall in mice, including endocardium, epicardium, and myocardium, and its expression was decreased with age. Importantly, the CD51+cMSCs possessed potent self-renewal potential and multi-lineage differentiation capacity in vitro and also expressed typical MSC-related surface proteins. Furthermore, CD51+cMSC transplantation significantly improved cardiac function and attenuated cardiac fibrosis through pro-angiogenesis activity after myocardial infarction in mice. Moreover, SCF secreted by CD51+cMSCs played an important role in angiogenesis both in vivo and in vitro. Conclusions: Collectively, CD51 is a novel marker of cardiac resident MSCs, and CD51+cMSC therapy enhances cardiac repair at least partly through SCF-mediated angiogenesis.

4.
Biomed Pharmacother ; 133: 110868, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33181455

RESUMEN

Male infertility has become an important health problem that is primarily caused by testicular dysfunction with abnormal spermatogenesis. In this study, we demonstrated that the neuropeptide, substance P (SP), is essential for spermatogonia proliferation in a seminiferous tubule culture system. In addition, SP (5 nmol/kg) treatment markedly restored spermatogenesis, improved sperm quality, and increased the number of ZBTB16+ or LIN28+ undifferentiated spermatogonia as well as STRA8+ differentiated spermatogonia in a busulfan-induced non-obstructive azoospermic mouse model. Furthermore, 100 nM SP treatment in vitro significantly stimulated the proliferation of GC-1 spg cells (a spermatogonia cell line) via activation of the Erk1/2 signaling pathway. Moreover, the sperm quality and the number of spermatogonia were significantly reduced after treatment with RP67580, a selective NK-1 receptor antagonist, suggesting that SP-NK1R signaling plays an important role in spermatogenesis. Taken together, these results suggest that SP may be a potential therapeutic agent for male infertility by accelerating the restoration of spermatogenesis.


Asunto(s)
Azoospermia/tratamiento farmacológico , Fármacos para la Fertilidad Masculina/farmacología , Fertilidad/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Sustancia P/farmacología , Animales , Apoptosis/efectos de los fármacos , Azoospermia/inducido químicamente , Azoospermia/metabolismo , Azoospermia/fisiopatología , Busulfano , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Receptores de Neuroquinina-1/agonistas , Receptores de Neuroquinina-1/metabolismo , Transducción de Señal , Espermatogonias/metabolismo , Técnicas de Cultivo de Tejidos
5.
Mol Ther ; 28(3): 855-873, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31991111

RESUMEN

Mesenchymal stromal cells (MSCs) show potential for treating cardiovascular diseases, but their therapeutic efficacy exhibits significant heterogeneity depending on the tissue of origin. This study sought to identify an optimal source of MSCs for cardiovascular disease therapy. We demonstrated that Nestin was a suitable marker for cardiac MSCs (Nes+cMSCs), which were identified by their self-renewal ability, tri-lineage differentiation potential, and expression of MSC markers. Furthermore, compared with bone marrow-derived MSCs (Nes+bmMSCs) or saline-treated myocardial infarction (MI) controls, intramyocardial injection of Nes+cMSCs significantly improved cardiac function and decreased infarct size after acute MI (AMI) through paracrine actions, rather than transdifferentiation into cardiac cells in infarcted heart. We further revealed that Nes+cMSC treatment notably reduced pan-macrophage infiltration while inducing macrophages toward an anti-inflammatory M2 phenotype in ischemic myocardium. Interestingly, Periostin, which was highly expressed in Nes+cMSCs, could promote the polarization of M2-subtype macrophages, and knockdown or neutralization of Periostin remarkably reduced the therapeutic effects of Nes+cMSCs by decreasing M2 macrophages at lesion sites. Thus, the present work systemically shows that Nes+cMSCs have greater efficacy than do Nes+bmMSCs for cardiac healing after AMI, and that this occurs at least partly through Periostin-mediated M2 macrophage polarization.


Asunto(s)
Moléculas de Adhesión Celular/genética , Activación de Macrófagos/genética , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Isquemia Miocárdica/etiología , Isquemia Miocárdica/metabolismo , Nestina/metabolismo , Cicatrización de Heridas/genética , Animales , Biomarcadores , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Genotipo , Inmunofenotipificación , Células Madre Mesenquimatosas/citología , Ratones , Ratones Transgénicos , Isquemia Miocárdica/patología
6.
Stem Cell Res Ther ; 10(1): 127, 2019 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-31029167

RESUMEN

BACKGROUND: Bone-derived mesenchymal stem cell (BMSC) transplantation has been reported to be effective for the treatment of ischemic heart disease, but whether BMSCs are the optimal cell type remains under debate. Increasing numbers of studies have shown that Nestin, an intermediate filament protein, is a potential marker for MSCs, which raises the question of whether Nestin+ cells in BMSCs may play a more crucial role in myocardial repair. METHODS: Nestin+ cells were isolated using flow cytometry by gating for CD45- Ter119- CD31- cells from the compact bone of Nestin-GFP transgenic mice, expressing GFP driven by the Nestin promoter. Colony-forming and proliferative curve assays were conducted to determine the proliferative capacity of these cells, while qRT-PCR was used to analyze the mRNA levels of relative chemokines and growth factors. Cardiac endothelial cell (CEC) recruitment was assessed via a transwell assay. Moreover, permanent ligation of the left anterior descending (LAD) coronary artery was performed to establish an acute myocardial infarction (AMI) mouse model. After cell transplantation, conventional echocardiography was conducted 1 and 4 weeks post-MI, and hearts were harvested for hematoxylin-and-eosin (HE) staining and immunofluorescence staining 1 week post-MI. Further evaluation of paracrine factor levels and administration of a neutralizing antibody (TIMP-1, TIMP-2, and CXCL12) or a CXCR4 antagonist (AMD3100) in MI hearts were performed to elucidate the mechanism involved in the chemotactic effect of Nestin+ BMSCs in vivo. RESULTS: Compared with Nestin- BMSCs, a greater proliferative capacity of Nestin+ BMSCs was observed, which further exhibited moderately high expression of chemokines instead of growth factors. More CEC recruitment in the Nestin+ BMSC-cocultured group was observed in vitro, while this effect was obviously abolished after treatment with neutralizing antibodies against TIMP-1, TIMP-2, or CXCL12, and more importantly, blocking the CXCL12/CXCR4 axis with a AMD3100 significantly reduced the chemotactic effect of Nestin+ BMSCs. After transplantation into mice exposed to myocardial infarction (MI), Nestin+ BMSC-treated mice showed significantly improved survival and left ventricular function compared with Nestin- BMSC-treated mice. Moreover, endogenous CECs were markedly increased, and chemokine levels were significantly higher, in the infarcted border zone with Nestin+ BMSC treatment. Meanwhile, neutralization of each TIMP-1, TIMP-2, or CXCL12 in vivo could reduce the left ventricular function at 1 and 4 weeks post-MI; importantly, the combined use of these three neutralizing antibodies could make a higher significance on cardiac function. Finally, blocking the CXCL12/CXCR4 axis with AMD3100 significantly reduced the left ventricular function and greatly inhibited Nestin+ BMSC-induced CEC chemotaxis in vivo. CONCLUSIONS: These results suggest that Nestin+ BMSC transplantation can improve cardiac function in an AMI model by recruiting resident CECs to the infarcted border region via the CXCL12/CXCR4 chemokine pathway. And we demonstrated that Nestin+BMSC-secreted TIMP-1/2 enhances CXCL12(SDF1α)/CXCR4 axis-driven migration of endogenous Sca-1+ endothelial cells in ischemic heart post-AMI. Taken together, our results show that Nestin is a useful marker for the identification of functional BMSCs and indicate that Nestin+ BMSCs could be a better therapeutic candidate for cardiac repair.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/terapia , Nestina/genética , Animales , Anticuerpos Neutralizantes/farmacología , Bencilaminas , Huesos/citología , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/genética , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Ciclamas , Células Endoteliales/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , ARN Mensajero/genética , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética , Inhibidor Tisular de Metaloproteinasa-1/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-2/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-2/genética
7.
Int Heart J ; 59(1): 180-189, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29375111

RESUMEN

Dilated cardiomyopathy (DCM), as one of the common cardiomyopathies, is a disease of the heart muscle; however, the etiology and pathogenesis of DCM were still poorly understood. Nestin has been reported a special marker of stem/progenitor cells in various tissues, and the tissue resident Nestin+ cells could promote the wound healing and tissue remodeling. However, it remains unclear whether Nestin+ cells participate in the protection of cardiomyocytes during the pathogenesis of DCM. Here the model of mice DCM was induced by doxorubicin (DOX) intraperitoneal injection and observed heart failure and ventricular enlargement via echocardiography and histologic analysis, respectively. During DCM pathogenesis, the number of Nestin+ cells showed a significant peak on day 6 after DOX treatment, which then gradually decreases to lower than normal levels after day 30 in the total population of the heart. Furthermore, we found that the isolated increased heart-derived Nestin+ cells are mesenchymal property and could protect DOX-induced HL-1 cells toxicity in vitro by promoting their proliferation and inhibiting their apoptosis. Collectively, our results showed that Nestin+ cells increased during DCM pathogenesis and played an important role in protecting against the DOX-induced HL-1 cells loss via regulating proliferation and apoptosis. Thus, the loss of Nestin+ cells might be an etiology to DCM pathogenesis, and these cells could be a promising candidate cell source for study and treatment of DCM patients.


Asunto(s)
Apoptosis , Cardiomiopatía Dilatada/genética , Regulación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Nestina/genética , ARN/genética , Función Ventricular Izquierda/fisiología , Animales , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Doxorrubicina/toxicidad , Ecocardiografía , Citometría de Flujo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/patología , Nestina/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Cell Death Dis ; 8(10): e3123, 2017 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29022899

RESUMEN

Previous studies have demonstrated that rodent stem Leydig cell (SLC) transplantation can partially restore testosterone production in Leydig cell (LC)-disrupted or senescent animal models, which provides a promising approach for the treatment of hypogonadism. Here, we isolated human SLCs prospectively and explored the potential therapeutic benefits of human SLC transplantation for hypogonadism treatment. In adult human testes, p75 neurotrophin receptor positive (p75+) cells expressed the known SLC marker nestin, but not the LC lineage marker hydroxysteroid dehydrogenase-3ß (HSD3ß). The p75+ cells which were sorted by flow cytometry from human adult testes could expand in vitro and exhibited clonogenic self-renewal capacity. The p75+ cells had multi-lineage differentiation potential into multiple mesodermal cell lineages and testosterone-producing LCs in vitro. After transplantation into the testes of ethane dimethane sulfonate (EDS)-treated LC-disrupted rat models, the p75+ cells differentiated into LCs in vivo and secreted testosterone in a physiological pattern. Moreover, p75+ cell transplantation accelerated the recovery of serum testosterone levels, spermatogenesis and reproductive organ weights. Taken together, we reported a method for the identification and isolation of human SLCs on the basis of p75 expression, and demonstrated that transplanted human p75+ SLCs could replace disrupted LCs for testosterone production. These findings provide the groundwork for further clinical application of human SLCs for hypogonadism.


Asunto(s)
Células Intersticiales del Testículo/metabolismo , Células Intersticiales del Testículo/trasplante , Proteínas del Tejido Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/genética , Trasplante de Células Madre , Células Madre/metabolismo , Testosterona/biosíntesis , Adolescente , Adulto , Animales , Biomarcadores/análisis , Células Cultivadas , Humanos , Hipogonadismo/terapia , Infertilidad Masculina/terapia , Masculino , Mesilatos/farmacología , Ratas , Ratas Sprague-Dawley , Espermatogénesis/fisiología , Trasplante Heterólogo , Adulto Joven
9.
J Sex Med ; 14(11): 1297-1306, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28943032

RESUMEN

BACKGROUND: Formaldehyde, a ubiquitous environmental pollutant, is used extensively and has been proved to impair male reproduction in mammals. However, no trials have explored whether formaldehyde affects sexual function. AIM: To evaluate the effect of long-term formaldehyde exposure on sexual behavior and to investigate the potential mechanism. METHODS: Forty C57BL/6 male mice were randomly allocated to four equally sized groups. Mice were exposed to formaldehyde at a dose of 0 (control), 0.5, 5.0, or 10.0 mg/m3 by inhalation for 60 days. OUTCOMES: Sexual behavior, body and reproductive organ weights, testosterone concentration in serum and testicular tissue, expression of steroidogenic enzymes, quality of sperm, and testicular structure were measured. RESULTS: Formaldehyde inhibited sexual behavior and decreased reproductive organ weights in mice. Serum testosterone levels and intratesticular testosterone concentrations were decreased in the formaldehyde-treated groups. Expression levels of steroidogenic enzymes, including steroidogenic acute regulatory protein, cytochrome P450 cholesterol side-chain cleavage enzyme, and 3ß-hydroxysteroid dehydrogenase (3ß-HSD), also were decreased in the testes of mice exposed to formaldehyde. Moreover, the structure of seminiferous tubules was destroyed and sperm quality decreased after formaldehyde exposure. In addition, the results indicated that the effects of formaldehyde were dose dependent. CLINICAL IMPLICATIONS: Efforts should be undertaken to decrease impairment of sexual function caused by formaldehyde exposure. STRENGTHS AND LIMITATIONS: The relatively small sample might have affected the outcomes. Further experiments are needed to study the mechanism of action of formaldehyde. CONCLUSION: Exposure to formaldehyde gas inhibited sexual behavior, caused reproductive organ atrophy, and impaired spermatogenesis in male mice, which might have been induced by suppressed expression of steroidogenic enzymes in Leydig cells and decreased testosterone synthesis. Zang Z-J, Fang Y-Q, Ji S-Y, et al. Formaldehyde Inhibits Sexual Behavior and Expression of Steroidogenic Enzymes in the Testes of Mice. J Sex Med 2017;14:1297-1306.


Asunto(s)
Formaldehído/farmacología , Fosfoproteínas/metabolismo , Conducta Sexual Animal/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Testículo/metabolismo , Animales , Células Intersticiales del Testículo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Testosterona/sangre
10.
Biomed Res Int ; 2017: 1252851, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28713821

RESUMEN

PURPOSE: The fate and function of the induced Schwann cells (iSCs) like cells from adipose tissue have not been critically evaluated in vivo after transplantation. The objective of this study is to compare the fate of iSCs with naïve SCs (nSCs) after transplantation into the lesion sites of sciatic nerve, respectively. METHODS: Adipose-derived stem cells from eGFP-expressing transgenic rat's subcutaneous fat were induced to iSCs in vitro. iSCs were injected to the sciatic nerve lesion area after crush injury and the cells fate was comparatively analyzed with that of nSCs from the same rat. RESULTS: At 12 weeks after transplantation, nSCs were detected only in the restricted area of cell transplantation site but iSCs were widely distributed all over the sciatic nerve. Based on double fluorescence observations, both iSCs and naïve ones were colocalized with P0-expressing myelin sheath, outbound by laminin-expressing basal membrane, and terminated at contactin-associated protein-expressing doublets. However, some of iSCs were also differentiated to the fibrocyte/fibroblast-like cells. In the histological analysis of repaired sciatic nerves, axon density was higher in iSC-received group than in the nSCs group and normal sciatic nerve. CONCLUSION: iSCs induced from subcutaneous fat tissues have higher engraftment and migration capacity than nSCs.


Asunto(s)
Adipocitos/trasplante , Células de Schwann/trasplante , Nervio Ciático/trasplante , Trasplante de Células Madre , Tejido Adiposo/citología , Tejido Adiposo/trasplante , Animales , Animales Modificados Genéticamente , Ratas , Células de Schwann/citología , Nervio Ciático/lesiones , Nervio Ciático/fisiopatología
11.
Int J Biochem Cell Biol ; 89: 110-119, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28579528

RESUMEN

Endogenous neural stem cells (NSCs) are the most promising sources for replacing cells lost after spinal cord injury (SCI). We have previously shown that substance P (SP), a neuropeptide, improves functional recovery after SCI and increases the numbers of cells in lesion sites, but how this occurs is unclear. Here, we investigate whether SP regulates the neurogenesis of resident NSCs as well as exerting a beneficial effect on functional improvement. We found that SP (5nmol/kg) treatment markedly improved functional recovery and elicited robust activation of endogenous NSCs and boosted the number of EdU+ proliferating cells differentiating into neurons, but it reduced astroglial differentiation in the lesion sites. Consistently, treatment with SP (10nM) in vitro significantly increased the proliferation of NSCs via activating the Erk1/2 signaling pathway and promoted neuronal differentiation but not astroglial differentiation. These results suggest that SP may represent a potential therapeutic agent for SCI via enhancing endogenous neurogenesis.


Asunto(s)
Neurogénesis/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Sustancia P/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Neuronas/efectos de los fármacos , Neuronas/patología
12.
Stem Cells ; 35(5): 1222-1232, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28090714

RESUMEN

Stem Leydig cell (SLC) transplantation could provide a new strategy for treating the testosterone deficiency. Our previous study demonstrated that CD51 (also called integrin αv) might be a putative cell surface marker for SLCs, but the physiological function and efficacy of CD51+ SLCs treatment remain unclear. Here, we explore the potential therapeutic benefits of CD51+ SLCs transplantation and whether these transplanted cells can be regulated by the hypothalamic-pituitary-gonadal (HPG) axis. CD51+ cells were isolated from the testes of 12-weeks-old C57BL/6 mice, and we showed that such cells expressed SLC markers and that they were capable of self-renewal, extensive proliferation, and differentiation into multiple mesenchymal cell lineages and LCs in vitro. As a specific cytotoxin that eliminates Leydig cells (LCs) in adult rats, ethane dimethanesulfonate (EDS) was used to ablate LCs before the SLC transplantation. After being transplanted into the testes of EDS-treated rats, the CD51+ cells differentiated into mature LCs, and the recipient rats showed a partial recovery of testosterone production and spermatogenesis. Notably, a testosterone analysis revealed a circadian rhythm of testosterone secretion in cell-transplanted rats, and these testosterone secretions could be suppressed by decapeptyl (a luteinizing hormone-releasing hormone agonist), suggesting that the transplanted cells might be regulated by the HPG axis. This study is the first to demonstrate that CD51+ SLCs can restore the neuroendocrine regulation of testicular function by physiologically recovering the expected episodic changes in diurnal testosterone serum levels and that SLC transplantation may provide a new tool for the studies of testosterone deficiency treatment. Stem Cells 2017;35:1222-1232.


Asunto(s)
Integrina alfaV/metabolismo , Células Intersticiales del Testículo/citología , Trasplante de Células Madre , Células Madre/citología , Testosterona/deficiencia , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Autorrenovación de las Células , Separación Celular , Modelos Animales de Enfermedad , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Mesilatos , Ratones Endogámicos C57BL , Tamaño de los Órganos , Ratas Sprague-Dawley , Espermatogénesis , Testículo/citología
13.
Sci Rep ; 6: 36995, 2016 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-27841342

RESUMEN

To evaluate the efficacy of magnetic resonance imaging (MRI) parameter optimizations for the diagnosis of periprosthetic infection and tumor recurrence in joint replacement patients. We compared the quality of images for 16 joint replacement patients that were recorded using the optimized MRI parameters with and without view angle tilting (VAT) correction at 1.5 T in coronal fast-spin-echo T2-weighted MRI. The optimized MRI data of 86 patients with pain after hip replacement and 67 patients who received tumor resection and joint replacement for bone cancer were retrospectively analyzed to identify MRI features that were useful for the diagnosis of periprosthetic infection and tumor recurrence. Increasing receiver bandwidth and decreasing slice thickness combined with VAT significantly reduced the area of metal-induced artifacts. Irregular soft tissue mass, soft tissue edema, bone destruction, and fistula were significant features of periprosthetic infection, with sensitivities of 47.4-100% and specificities of 73.1-100.0%, which were confirmed based on surgical and pathological findings. Soft tissue mass was a significant feature of tumor recurrence, with 100% sensitivity, 96.0% specificity, and 97.0% consistency. The optimized VAT MRI method demonstrated a high level of diagnostic accuracy for the detection of periprosthetic infection and tumor recurrence in joint replacement patients.


Asunto(s)
Neoplasias Óseas/patología , Imagen por Resonancia Magnética , Infecciones Relacionadas con Prótesis/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Aleaciones/química , Artroplastia de Reemplazo de Cadera , Artefactos , Neoplasias Óseas/cirugía , Femenino , Humanos , Interpretación de Imagen Asistida por Computador , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Prótesis e Implantes , Estudios Retrospectivos , Sensibilidad y Especificidad
14.
Mol Ther ; 24(10): 1860-1872, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27397633

RESUMEN

Mesenchymal stromal cells (MSCs) have shown great potential for treating inflammatory bowel disease, which is ameliorated through paracrine cross talk between MSCs and T-cells. Members of the insulin-like growth factor binding protein (IGFBP) family have important immunomodulatory functions in MSCs, but the underlying mechanisms behind these functions have not yet been clearly elucidated. In this study, we investigate whether MSC-produced IGFBP7 is involved in immune modulation using a mouse experimental colitis model. Gene expression profiling revealed that IGFBP7 was highly expressed in MSCs. Consistent with this findings, IGFBP7 knockdown in MSCs significantly decreased their immunomodulatory properties, decreasing the antiproliferative functions of MSCs against T-cells, while also having an effect on the proinflammatory cytokine production of the T-cells. Furthermore, in the mouse experimental colitis model, MSC-derived IGFBP7 ameliorated the clinical and histopathological severity of induced colonic inflammation and also restored the injured gastrointestinal mucosal tissues. In conclusion, IGFBP7 contributes significantly to MSC-mediated immune modulation, as is shown by the ability of IGFBP7 knockdown in MSCs to restore proliferation and cytokine production in T-cells. These results suggest that IGFBP7 may act as a novel MSC-secreted immunomodulatory factor.


Asunto(s)
Colitis/terapia , Factores Inmunológicos/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Colitis/inducido químicamente , Colitis/metabolismo , Modelos Animales de Enfermedad , Células Madre Mesenquimatosas/metabolismo , Ratones , Regulación hacia Arriba
15.
Biomaterials ; 50: 56-66, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25736496

RESUMEN

Renal resident mesenchymal stem cells (MSCs) are important regulators of kidney homeostasis, repair or regeneration. However, natural distribution and the starting population properties of these cells remain elusive because of the lack of specific markers. Here, we identified post-natal kidney derived Nestin(+) cells that fulfilled all of the criteria as a mesenchymal stem cell. These isolated Nestin(+) cells expressed the typical cell-surface marker of MSC, including Sca-1, CD44, CD106, NG2 and PDGFR-α. They were capable of self-renewal, possessed high clonogenic potential and extensive proliferation for more than 30 passages. Under appropriate differentiation conditions, these cells could differentiate into adipocytes, osteocytes, chondrocytes and podocytes. After intravenous injection into acute kidney injury mice, Nestin(+) cells contributed to functional improvement by significantly decreasing the peak level of serum creatinine and BUN, and reducing the damaged cell apoptosis. Furthermore, conditioned medium from Nestin(+) cells could protect against ischemic acute renal failure partially through paracrine factor VEGF. Taken together, our findings indicate that renal resident Nestin(+) MSCs can be derived, propagated, differentiated, and repair the acute kidney injury, which may shed new light on understanding MSCs biology and developing cell replacement therapies for kidney disease.


Asunto(s)
Lesión Renal Aguda/terapia , Isquemia/terapia , Riñón/irrigación sanguínea , Riñón/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Nestina/metabolismo , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Separación Celular , Células Clonales , Ensayo de Unidades Formadoras de Colonias , Medios de Cultivo Condicionados/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Isquemia/complicaciones , Isquemia/patología , Isquemia/fisiopatología , Riñón/metabolismo , Riñón/fisiopatología , Pruebas de Función Renal , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Sustancias Protectoras/metabolismo , Recuperación de la Función/efectos de los fármacos , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Cell Res ; 24(12): 1466-85, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25418539

RESUMEN

The ability to identify and isolate lineage-specific stem cells from adult tissues could facilitate cell replacement therapy. Leydig cells (LCs) are the primary source of androgen in the mammalian testis, and the prospective identification of stem Leydig cells (SLCs) may offer new opportunities for treating testosterone deficiency. Here, in a transgenic mouse model expressing GFP driven by the Nestin (Nes) promoter, we observed Nes-GFP+ cells located in the testicular interstitial compartment where SLCs normally reside. We showed that these Nes-GFP+ cells expressed LIFR and PDGFR-α, but not LC lineage markers. We further observed that these cells were capable of clonogenic self-renewal and extensive proliferation in vitro and could differentiate into neural or mesenchymal cell lineages, as well as LCs, with the ability to produce testosterone, under defined conditions. Moreover, when transplanted into the testes of LC-disrupted or aging models, the Nes-GFP+ cells colonized the interstitium and partially increased testosterone production, and then accelerated meiotic and post-meiotic germ cell recovery. In addition, we further demonstrated that CD51 might be a putative cell surface marker for SLCs, similar with Nestin. Taken together, these results suggest that Nes-GFP+ cells from the testis have the characteristics of SLCs, and our study would shed new light on developing stem cell replacement therapy for testosterone deficiency.


Asunto(s)
Células Madre Adultas/trasplante , Células Intersticiales del Testículo/patología , Células Intersticiales del Testículo/trasplante , Nestina/análisis , Testículo/citología , Testículo/patología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Envejecimiento , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Expresión Génica , Integrina alfaV/análisis , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Nestina/genética , Estudios Prospectivos , Espermatogénesis , Testículo/fisiología , Testosterona/metabolismo
17.
Neuroreport ; 24(15): 846-51, 2013 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23995292

RESUMEN

Previously, we have reported that substance P (SP) enhanced functional recovery from spinal cord injury (SCI) possibly by the anti-inflammatory modulation associated with the induction of M2-type macrophages at the injured lesion. In this study, we explored the cytokine expression profiles and apoptotic cell death in the lesion site of the SCI after an immediate intravenous injection of SP. SP injection increased the levels of interleukin-4 (IL-4), IL-6, and IL-10 at day 1 after the SCI approximately by 2-, 9-, and 10-folds when compared with the control SCI, respectively. On the basis of double immunofluorescence staining with IL-10 and CD11b, activated macrophages or microglia expressing IL-10 appeared in the margin of the lesion site at day 1 only after the SP injection. This SP-mediated alteration in the lesion microenvironment was shown to be associated with the lower cell death of neuronal cells at day 1 and oligodendrocytes at day 5 by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, which was also accompanied by a decrease in caspase-3 activation. These findings suggest that SP may reduce the inflammation-induced secondary cell death, possibly through immune modulation at an early stage after the SCI.


Asunto(s)
Antiinflamatorios/farmacología , Apoptosis , Citocinas/metabolismo , Traumatismos de la Médula Espinal/inmunología , Sustancia P/farmacología , Animales , Antiinflamatorios/administración & dosificación , Inflamación/inmunología , Masculino , Ratas , Ratas Sprague-Dawley , Sustancia P/administración & dosificación , Factores de Tiempo
18.
Zhonghua Nan Ke Xue ; 19(5): 392-7, 2013 May.
Artículo en Chino | MEDLINE | ID: mdl-23757958

RESUMEN

OBJECTIVE: To explore the P75NTR expression in the mouse testis and its relationship with nestin. METHODS: We observed the location of the expressions of P75NTR and nestin in the testis of the nestin-GFP transgenic mouse on postnatal day (PND) 5, 14 and 30 using immunofluorescence, and detected the expression levels of P75NTR in the testicular tissue of mice in different age groups by real-time quantitative PCR (RTqPCR) and flow cytometry. Then we cultured the P75NTR positive cells in neural stem cell culture medium and observed their neuronal differentiation capacity by orientation differentiation. RESULTS: Immunofluorescence showed the expressions of P75NTR and nestin in the Leydig cells of the mouse testis. RTqPCR and flow cytometry exhibited the peak of the P75NTR expression on PND 14. The positive rates of P75NTR were (2.88 +/- 0.52), (9.54 +/- 1.81) and (2.63 +/- 0.43)% on PND 5, 14 and 30, respectively. The P75NTR positive cells obtained also expressed nestin and P75NTR and had the capacity of neuronal differentiation. CONCLUSION: P75NTR and nestin are co-expressed in the Leydig cells of the mouse testis, and the P75NTR positive cells have the ability of neural differentiation, which is presumably attributed to neural crest cells.


Asunto(s)
Proteínas de Filamentos Intermediarios/genética , Células Intersticiales del Testículo/metabolismo , Proteínas del Tejido Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/genética , Animales , Proteínas de Filamentos Intermediarios/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Nestina , Receptor de Factor de Crecimiento Nervioso/metabolismo , Testículo/citología , Testículo/metabolismo
19.
Biomaterials ; 34(29): 7086-96, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23791502

RESUMEN

Introducing concentration gradients of nerve growth factor (NGF) into conduits for repairing of peripheral nerve injury is crucial for nerve regeneration and guidance. Herein, combining differential adsorption of NGF/silk fibroin (SF) coating, the gradient of NGF-immobilized membranes (G-Ms) and nanofibrous nerve conduits (G-nNCs) were successfully fabricated. The efficacy of NGF gradients was confirmed by a quantitative comparison of dorsal root ganglia (DRG) neurite outgrowth on the G-Ms or uniform NGF-immobilized membranes (U-Ms). Significantly, the neurite turning ratio was 0.48 ± 0.11 for G-M group, but it was close to zero for U-M group. The neurite length of DRGs in the middle of the G-Ms was significantly longer than that of U-M group, even though the average NGF concentration was approximated. Furthermore, 12 weeks after implantation in rats with a 14 mm gap of sciatic nerve injury, G-nNCs achieved satisfying outcomes of nerve regeneration associated with morphological and functional improvements, which was superior to that of the uniform NGF-immobilized nNCs (U-nNCs). Sciatic function index (SFI), compound muscle action potentials (CMAPs), total number of myelinated nerve fibers, thickness of myelin sheath were similar for the G-nNCs and autografts, with the G-nNCs having a higher density of axons than the autografts. Our results demonstrated the significant role of introducing NGF gradients into scaffolds in promoting nerve regeneration.


Asunto(s)
Ganglios Espinales/fisiología , Factor de Crecimiento Nervioso/administración & dosificación , Regeneración Nerviosa/efectos de los fármacos , Nervio Ciático/fisiología , Andamios del Tejido , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Ganglios Espinales/citología , Regeneración Tisular Dirigida , Masculino , Membranas Artificiales , Factor de Crecimiento Nervioso/uso terapéutico , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Ciático/efectos de los fármacos , Nervio Ciático/lesiones , Andamios del Tejido/química
20.
Neuroreport ; 22(12): 603-8, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21753712

RESUMEN

We induced human melanocyte dedifferentiation to Schwann cell-like cells in vitro by a combination of forskolin, neuregulin-ß1, neurotrophin-3, platelet-derived growth factor-aa, basic fibroblast growth factor, laminin, and heparin. Cultured human melanocytes constitutively expressed neural cell and melanocyte markers but melanocyte-specific marker, including microphthalmia-associated transcription factor and tyrosinase, expression was selectively lost after induction. In the sciatic nerve injury site, the induced cells were engrafted and closely aligned to axons and P0-expressing myelin sheaths, whereas uninduced cells were not colocalized with axons and myelin sheaths and reexpressed melanocyte-specific tyrosinase activity in vivo. Human melanocytes lose their melanocyte phenotype and transdifferentiate into Schwann cells under specific induction conditions and display their Schwann cell phenotype after transplantation to injured sciatic nerve tissue.


Asunto(s)
Transdiferenciación Celular/fisiología , Melanocitos/citología , Regeneración Nerviosa/fisiología , Células de Schwann/citología , Células de Schwann/trasplante , Nervio Ciático/cirugía , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nervio Ciático/lesiones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...