Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Reprod Toxicol ; 126: 108584, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38561096

RESUMEN

In the domain of medical advancement, nanotechnology plays a pivotal role, especially in the synthesis of biocompatible materials for therapeutic use. Superparamagnetic Iron Oxide Nanoparticles (SPIONs), known for their magnetic properties and low toxicity, stand at the forefront of this innovation. This study explored the reproductive toxicological effects of Sodium Citrate-functionalized SPIONs (Cit_SPIONs) in adult male mice, an area of research that holds significant potential yet remains largely unknown. Our findings reveal that Cit_SPIONs induce notable morphological changes in interstitial cells and the seminiferous epithelium when introduced via intratesticular injection. This observation is critical in understanding the interactions of nanomaterials within reproductive biological systems. A striking feature of this study is the rapid localization of Cit_SPIONs in Leydig cells post-injection, a factor that appears to be closely linked with the observed decrease in steroidogenic activity and testosterone levels. This data suggests a possible application in developing nanostructured therapies targeting androgen-related processes. Over 56 days, these nanoparticles exhibited remarkable biological distribution in testis parenchyma, infiltrating various cells within the tubular and intertubular compartments. While the duration of spermatogenesis remained unchanged, there were many Tunel-positive germ cells, a notable reduction in daily sperm production, and reduced progressive sperm motility in the treated group. These insights not only shed light on the intricate mechanisms of Cit_SPIONs interaction with the male reproductive system but also highlight the potential of nanotechnology in developing advanced biomedical applications.

2.
BMC Biol ; 21(1): 36, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36797789

RESUMEN

BACKGROUND: Cellular entry of SARS-CoV-2 has been shown to rely on angiotensin-converting enzyme 2 (ACE2) receptors, whose expression in the testis is among the highest in the body. Additionally, the risk of mortality seems higher among male COVID-19 patients, and though much has been published since the first cases of COVID-19, there remain unanswered questions regarding SARS-CoV-2 impact on testes and potential consequences for reproductive health. We investigated testicular alterations in non-vaccinated deceased COVID-19-patients, the precise location of the virus, its replicative activity, and the immune, vascular, and molecular fluctuations involved in the pathogenesis. RESULTS: We found that SARS-CoV-2 testicular tropism is higher than previously thought and that reliable viral detection in the testis requires sensitive nanosensors or RT-qPCR using a specific methodology. Through an in vitro experiment exposing VERO cells to testicular macerates, we observed viral content in all samples, and the subgenomic RNA's presence reinforced the replicative activity of SARS-CoV-2 in testes of the severe COVID-19 patients. The cellular structures and viral particles, observed by transmission electron microscopy, indicated that macrophages and spermatogonial cells are the main SARS-CoV-2 lodging sites, where new virions form inside the endoplasmic reticulum Golgi intermediate complex. Moreover, we showed infiltrative infected monocytes migrating into the testicular parenchyma. SARS-CoV-2 maintains its replicative and infective abilities long after the patient's infection. Further, we demonstrated high levels of angiotensin II and activated immune cells in the testes of deceased patients. The infected testes show thickening of the tunica propria, germ cell apoptosis, Sertoli cell barrier loss, evident hemorrhage, angiogenesis, Leydig cell inhibition, inflammation, and fibrosis. CONCLUSIONS: Our findings indicate that high angiotensin II levels and activation of mast cells and macrophages may be critical for testicular pathogenesis. Importantly, our findings suggest that patients who become critically ill may exhibit severe alterations and harbor the active virus in the testes.


Asunto(s)
COVID-19 , Testículo , Tropismo Viral , Animales , Humanos , Masculino , Angiotensina II/metabolismo , Chlorocebus aethiops , COVID-19/patología , SARS-CoV-2 , Testículo/inmunología , Testículo/virología , Células Vero
3.
Toxicology ; 455: 152761, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33766575

RESUMEN

Several studies have reported the effects of atrazine on the gonads of many experimental models. However, the short-term effects of in vivo exposure to atrazine on the testes of mice are not well clarified. Here we reported that adult BalB/c mice exposed to atrazine (50 mg kg-1 body weight) by gavage for three consecutive days have reduced numbers of 3ß-hydroxysteroid dehydrogenase positive Leydig cells (LCs), associated with increased in situ cell death fluorescence and caspase-3 immuno-expression in the testes. Consequently, immunostaining for cell cycle gene regulators showed increased expressions of p45, accompanied with increased expressions of cyclin D2 and E2. Histological observations of the gonads showed reduced number of germ cells in particular areas, sloughed seminiferous epithelium, presence of giant apoptotic cells close to the seminiferous tubule lumen and in the epididymal lumen along with low numbers of Leydig cells in the testicular interstitial areas. Similarly, LCs isolated from the testes of BalB/c mice that were exposed to atrazine (0.5, 25, 50 mg kg-1 body weight) in the same manner as in the first experiment presented dose-dependent increased caspase-3 activity, decreased cell viability, intratesticular and serum testosterone concentrations and LCs testosterone secretion. In summary, atrazine appears to directly decrease the number of testosterone secreting LCs in mice through apoptosis.


Asunto(s)
Atrazina/toxicidad , Herbicidas/toxicidad , Células Intersticiales del Testículo/efectos de los fármacos , Testosterona/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Atrazina/administración & dosificación , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Herbicidas/administración & dosificación , Células Intersticiales del Testículo/patología , Masculino , Ratones , Ratones Endogámicos BALB C
4.
J Inorg Biochem ; 206: 111017, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32120160

RESUMEN

Cancer-Associated Fibroblasts (CAFs) contribute to tumour progression and have received significant attention as a therapeutic target. These cells produce growth factors, cytokines and chemokines, stimulating cancer cell proliferation and inhibiting their apoptosis. Recent advances in drug delivery have demonstrated a significant promise of iron oxide nanoparticles in clinics as theranostic agents, mainly due to their magnetic properties. Here, we designed superparamagnetic iron oxide nanoparticles (SPIONs) to induce apoptosis of human fibroblasts. SPIONs were synthesized via co-precipitation method and coated with sodium citrate (SPION_Cit). We assessed the intracellular uptake of SPIONs by human fibroblast cells, as well as their cytotoxicity and ability to induce thermal effects under the magnetic field. The efficiency and time of nanoparticle internalization were assessed by Prussian Blue staining, flow cytometry and transmission electron microscopy. SPIONs_Cit were detected in the cytoplasm of human fibroblasts 15 min after in vitro exposure, entering into cells mainly via endocytosis. Analyses through Cell Titer Blue assay, AnnexinV-fluorescein isothiocyanate (FITC) and propidium iodide (PI) cellular staining demonstrated that concentrations below 8 × 10-2 mg/mL of SPIONs_Cit did not alter cell viability of human fibroblast. Furthermore, it was also demonstrated that SPIONs_Cit associated with alternating current magnetic field were able to induce hyperthermia and human fibroblast cell death in vitro, mainly through apoptosis (83.5%), activating caspase 8 (extrinsic apoptotic via) after a short exposure period. Collectively these findings suggest that our nanoplatform is biocompatible and can be used for therapeutic purposes in human biological systems, such as inducing apoptosis of CAFs.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos Férricos/farmacología , Fibroblastos/efectos de los fármacos , Nanopartículas Magnéticas de Óxido de Hierro/administración & dosificación , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ácido Cítrico/química , Endocitosis , Compuestos Férricos/química , Citometría de Flujo , Humanos , Hipertermia Inducida , Nanopartículas Magnéticas de Óxido de Hierro/química , Microscopía Electrónica de Transmisión , Neoplasias/metabolismo , Neoplasias/patología
5.
J Therm Biol ; 77: 14-23, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30196893

RESUMEN

Temperature is considered a crucial modulator of reproductive activity and testis homeostasis. It is well known that elevated temperatures cause several effects on testicular components, particularly on germ cells, which might lead to the impairment of spermatogenesis and loss of male fertility. The present study aimed to evaluate the effects of different environmental temperatures on several morphofunctional testis parameters, with emphasis on duration of spermatogenesis and spermatogenic efficiency. Thirty sexually mature Swiss mice (Mus musculus) were allocated in three different experimental groups, being kept in vivarium for three weeks at 16 °C, 23 °C (control group) and 32 °C. In order to estimate the duration of spermatogenesis, three animals per each group received intraperitoneal injections of tritiated thymidine and the testes were perfused-fixed and routinely processed for histological, morphometrical and immunoperoxidase analyses. Although the lower temperature (16 °C) did not change most of the evaluated testicular parameters, our findings showed that higher environmental temperature (32 °C) is able to alter important testis parameters, resulting for instance in acceleration of spermatogenesis, alterations in the stages frequencies, increased number of germ and Leydig cells apoptosis and reduced Sertoli cell and spermatogenic efficiencies. As in many conditions infertile men exhibit higher mean scrotal temperature, we believe that experimental studies with mice involving temperature might represent an interesting approach to better understand the mechanisms related to human testis function and sperm production.


Asunto(s)
Espermatogénesis , Testículo/fisiología , Termotolerancia , Animales , Apoptosis , Temperatura Corporal , Calor , Infertilidad Masculina , Masculino , Ratones , Células de Sertoli/citología , Células de Sertoli/metabolismo , Células de Sertoli/ultraestructura , Espermatocitos/citología , Espermatocitos/metabolismo , Espermatozoides/citología , Espermatozoides/fisiología , Espermatozoides/ultraestructura , Testículo/citología , Testículo/ultraestructura , Testosterona/sangre
6.
Cell Tissue Res ; 370(3): 335-346, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28779347

RESUMEN

In recent decades, infertility has been considered a major widespread public health issue of very high concern. Currently, almost 50% of infertility cases are due to male factors, including semen disorders, obstructions, cryptorchidism, varicocele and testicular failures, which can occur due to malfunctions in both somatic and germ cells. In this context, besides other approaches, different miRNAs have been used as biomarkers for the diagnosis of male infertility, with different pathologic conditions such as Sertoli cell-only syndrome, mixed atrophy, and germ cell arrest. However, most studies related to male fertility do not point out the functions and cell targets of the described miRNAs. Initial investigations using experimental assays in murine and porcine models were performed, providing the first evidence of the influence of miRNAs on Sertoli cell function including, for instance, proliferation, maturation and hormone responses of these cells. The aim of this mini-review is therefore to summarize our present knowledge of this relevant subject and to highlight the importance of future investigations concerning the miRNA influence in the control of Sertoli cells, spermatogenesis and male fertility.


Asunto(s)
Infertilidad Masculina/genética , MicroARNs/genética , Células de Sertoli/metabolismo , Espermatogénesis/genética , Animales , Proliferación Celular/genética , ARN Helicasas DEAD-box/genética , Marcadores Genéticos/genética , Humanos , Masculino , Ribonucleasa III/genética , Síndrome de Sólo Células de Sertoli/diagnóstico , Síndrome de Sólo Células de Sertoli/genética , Espermatogénesis/fisiología , Porcinos , Testículo/fisiopatología
7.
Cell Tissue Res ; 370(3): 489-500, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28831567

RESUMEN

The establishment of proper conditions for spermatogonial stem cells (SSCs) cryopreservation and storage represents an important biotechnological approach for the preservation of the genetic stock of valuable animals. This study demonstrates the effects of different cryopreservation protocols on the survival rates and phenotypic expression of SSCs in horses. The cells were enzymatically isolated from testes of eight adult horses. After enrichment and characterization of germ cells in the suspension, the feasibility of several cryopreservation protocols were evaluated. Three different cryomedia compositions, associated with three different methods of freezing (vitrification, slow-freezing and fast-freezing) were evaluated. Based on the rates of viable SSCs found before and after thawing, as well as the number of recovered cells after cryopreservation, the best results were obtained utilizing the DMSO-based cryomedia associated with the slow-freezing method. In addition, when isolated cells were cultured in vitro, MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay and immunofluorescence analysis indicated that the cryopreserved cells were as metabolically active as the fresh cells and were also expressing typical SSCs proteins (VASA, NANOS2 and GFRA1). Therefore, our results indicate that equine SSCs can be cryopreserved without impairment of structure, function, or colony-forming abilities.


Asunto(s)
Células Madre Germinales Adultas/citología , Criopreservación/métodos , Preservación de Semen/métodos , Espermatogonias/citología , Vitrificación , Animales , Supervivencia Celular , Crioprotectores/farmacología , Dimetilsulfóxido/farmacología , Caballos , Masculino , Tejido Parenquimatoso/citología , Testículo/citología
8.
Biotechnol Adv ; 35(6): 832-844, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28602961

RESUMEN

The history of transgenesis is marked by milestones such as the development of cellular transdifferentiation, recombinant DNA, genetic modification of target cells, and finally, the generation of simpler genetically modified organisms (e.g. bacteria and mice). The first transgenic fish was developed in 1984, and since then, continuing technological advancements to improve gene transfer have led to more rapid, accurate, and efficient generation of transgenic animals. Among the established methods are microinjection, electroporation, lipofection, viral vectors, and gene targeting. Here, we review the history of animal transgenesis, with an emphasis on fish, in conjunction with major developments in genetic engineering over the past few decades. Importantly, spermatogonial stem cell modification and transplantation are two common techniques capable of revolutionizing the generation of transgenic fish. Furthermore, we discuss recent progress and future biotechnological prospects of fish transgenesis, which has strong applications for the aquaculture industry. Indeed, some transgenic fish are already available in the current market, validating continued efforts to improve economically important species with biotechnological advancements.


Asunto(s)
Animales Modificados Genéticamente/genética , Peces/genética , Técnicas de Transferencia de Gen/tendencias , Animales , Acuicultura/tendencias
9.
Cytometry A ; 85(1): 43-77, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24700575

RESUMEN

Stem cells are known for their capacity to self-renew and differentiate into at least one specialized cell type. Mesenchymal stem cells (MSCs) were isolated initially from bone marrow but are now known to exist in all vascularized organ or tissue in adults. MSCs are particularly relevant for therapy due to their simplicity of isolation and cultivation. The International Society for Cellular Therapy (ISCT) has proposed a set of standards to define hMSCs for laboratory investigations and preclinical studies: adherence to plastic in standard culture conditions; in vitro differentiation into osteoblasts, adipocytes, and chondroblasts; specific surface antigen expression in which ≥95% of the cells express the antigens recognized by CD105, CD73, and CD90, with the same cells lacking (≤2% positive) the antigens CD45, CD34, CD14 or CD11b, CD79a or CD19, and HLA-DR. In this review we will take an historical overview of how umbilical cord blood, bone marrow, adipose-derived, placental and amniotic fluid, and menstrual blood stem cells, the major sources of human MSC, can be obtained, identified and how they are being used in clinical trials to cure and treat a very broad range of conditions, including heart, hepatic, and neurodegenerative diseases. An overview of protocols for differentiation into hepatocytes, cardiomyocytes, neuronal, adipose, chondrocytes, and osteoblast cells are highlighted. We also discuss a new source of stem cells, induced pluripotent stem cells (iPS cells) and some pathways, which are common to MSCs in maintaining their pluripotent state.


Asunto(s)
Células Madre Adultas/inmunología , Diferenciación Celular/inmunología , Inmunofenotipificación , Osteoblastos/inmunología , Adipocitos/inmunología , Adulto , Antígenos CD/inmunología , Células de la Médula Ósea/inmunología , Condrocitos/inmunología , Humanos , Miocitos Cardíacos/inmunología
10.
PLoS One ; 7(8): e44091, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22937157

RESUMEN

Spermatogonial stem cells (SSCs) are the foundation of spermatogenesis and are located in a highly dynamic microenvironment called "niche" that influences all aspects of stem cell function, including homing, self-renewal and differentiation. Several studies have recently identified specific proteins that regulate the fate of SSCs. These studies also aimed at identifying surface markers that would facilitate the isolation of these cells in different vertebrate species. The present study is the first to investigate SSC physiology and niche in stallions and to offer a comparative evaluation of undifferentiated type A spermatogonia (Aund) markers (GFRA1, PLZF and CSF1R) in three different domestic equid species (stallions, donkeys, and mules). Aund were first characterized according to their morphology and expression of the GFRA1 receptor. Our findings strongly suggest that in stallions these cells were preferentially located in the areas facing the interstitium, particularly those nearby blood vessels. This distribution is similar to what has been observed in other vertebrate species. In addition, all three Aund markers were expressed in the equid species evaluated in this study. These markers have been well characterized in other mammalian species, which suggests that the molecular mechanisms that maintain the niche and Aund/SSCs physiology are conserved among mammals. We hope that our findings will help future studies needing isolation and cryopreservation of equids SSCs. In addition, our data will be very useful for studies that aim at preserving the germplasm of valuable animals, and involve germ cell transplantation or xenografts of equids testis fragments/germ cells suspensions.


Asunto(s)
Equidae/fisiología , Espermatogénesis/fisiología , Espermatogonias/citología , Nicho de Células Madre/fisiología , Células Madre/citología , Animales , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Espermatogonias/metabolismo , Células Madre/metabolismo , Testículo/citología , Testículo/metabolismo
11.
Biol Reprod ; 86(5): 155, 1-10, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22262689

RESUMEN

In the seminiferous epithelium, spermatogonial stem cells (SSCs) are located in a particular environment called the "niche" that is controlled by the basement membrane, key testis somatic cells, and factors originating from the vascular network. However, the role of Leydig cells (LCs) as a niche component is not yet clearly elucidated. Recent studies showed that peccaries (Tayassu tajacu) present a peculiar LC cytoarchitecture in which these cells are located around the seminiferous tubule lobes, making the peccary a unique model for investigating the SSC niche. This peculiarity allowed us to subdivide the seminiferous tubule cross-sections in three different testis parenchyma regions (tubule-tubule, tubule-interstitium, and tubule-LC contact). Our aims were to characterize the different spermatogonial cell types and to determine the location and/or distribution of the SSCs along the seminiferous tubules. Compared to differentiating spermatogonia, undifferentiated spermatogonia (A(und)) presented a noticeably higher nuclear volume (P < 0.05), allowing an accurate evaluation of their distribution. Immunostaining analysis demonstrated that approximately 93% of A(und) were GDNF receptor alpha 1 positive (GFRA1(+)), and these cells were preferentially located adjacent to the interstitial compartment without LCs (P < 0.05). The expression of colony-stimulating factor 1 was observed in LCs and peritubular myoid cells (PMCs), whereas its receptor was present in LCs and in GFRA1(+) A(und). Taken together, our findings strongly suggest that LCs, different from PMCs, might play a minor role in the SSC niche and physiology and that these steroidogenic cells are probably involved in the differentiation of A(und) toward type A(1) spermatogonia.


Asunto(s)
Espermatogonias/metabolismo , Nicho de Células Madre/fisiología , Animales , Artiodáctilos/fisiología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/análisis , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Masculino , Receptor de Factor Estimulante de Colonias de Macrófagos/análisis , Túbulos Seminíferos/citología , Espermatogénesis/fisiología , Espermatogonias/citología , Células Madre/citología , Células Madre/metabolismo
12.
J Androl ; 33(2): 264-76, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21597091

RESUMEN

With the exception of the domestic cat, all members of the family Felidae are considered either endangered or threatened. Although not yet used for this purpose, spermatogonial stem cell (SSC) transplantation has a high potential to preserve the genetic stock of endangered species. However, this technique has not previously been established in felids. Therefore, we developed the necessary procedures to perform syngeneic and xenogeneic SSC transplants (eg, germ cell [GC] depletion in the recipient domestic cats, enrichment and labeling of donor cell suspension, and the transplantation method) in order to investigate the feasibility of the domestic cat as a recipient for the preservation and propagation of male germ plasm from wild felids. In comparison with busulfan treatment, local x-ray fractionated radiation was a more effective approach to depleting endogenous spermatogenesis. The results of both syngeneic and xenogeneic transplants revealed that SSCs were able to successfully colonize and differentiate in the recipient testis, generating elongated spermatids several weeks posttransplantation. Specifically, ocelot spermatozoa were observed in the cat epididymis 13 weeks following transplantation. As donor GCs from domestic cats and ocelots were able to develop and form mature GCs in the recipient environment seminiferous tubules, these findings indicate that the domestic cat is a suitable recipient for SSC transplantation. Moreover, as modern cats descended from a medium-size cat that existed approximately 10 to 11 million years ago, these results strongly suggest that the domestic cat could be potentially used as a recipient for generating and propagating the genome of wild felids.


Asunto(s)
Especies en Peligro de Extinción , Felidae , Técnicas Reproductivas Asistidas/veterinaria , Espermatogonias/trasplante , Trasplante de Células Madre/veterinaria , Testículo/cirugía , Animales , Busulfano/farmacología , Gatos , Diferenciación Celular , Supervivencia Celular , Rastreo Celular , Fraccionamiento de la Dosis de Radiación , Estudios de Factibilidad , Masculino , Análisis de Semen , Espermatogénesis/efectos de los fármacos , Espermatogénesis/efectos de la radiación , Espermatogonias/efectos de los fármacos , Espermatogonias/efectos de la radiación , Factores de Tiempo , Trasplante Heterólogo , Trasplante Isogénico
13.
PLoS One ; 5(5): e10740, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20505774

RESUMEN

BACKGROUND: Germ cell transplantation results in fertile recipients and is the only available approach to functionally investigate the spermatogonial stem cell biology in mammals and probably in other vertebrates. In the current study, we describe a novel non-surgical methodology for efficient spermatogonial transplantation into the testes of adult tilapia (O. niloticus), in which endogenous spermatogenesis had been depleted with the cytostatic drug busulfan. METHODOLOGY/PRINCIPAL FINDINGS: Using two different tilapia strains, the production of fertile spermatozoa with donor characteristics was demonstrated in adult recipient, which also sired progeny with the donor genotype. Also, after cryopreservation tilapia spermatogonial cells were able to differentiate to spermatozoa in the testes of recipient fishes. These findings indicate that injecting germ cells directly into adult testis facilitates and enable fast generation of donor spermatogenesis and offspring compared to previously described methods. CONCLUSION: Therefore, a new suitable methodology for biotechnological investigations in aquaculture was established, with a high potential to improve the production of commercially valuable fish, generate transgenic animals and preserve endangered fish species.


Asunto(s)
Envejecimiento/fisiología , Cruzamiento/métodos , Cíclidos/fisiología , Modelos Biológicos , Reproducción/fisiología , Espermatogonias/trasplante , Animales , Animales Modificados Genéticamente , Busulfano , Criopreservación , Masculino , Filogenia , Espermatogonias/citología , Testículo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA