Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Biol Cell ; 116(6): e2300127, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38593304

RESUMEN

BACKGROUND: Spermatogenesis is a fundamental process crucial for male reproductive health and fertility. Exosomes, small membranous vesicles released by various cell types, have recently garnered attention for their role in intercellular communication. OBJECTIVE: This review aims to comprehensively explore the role of exosomes in regulating spermatogenesis, focusing on their involvement in testicular development and cell-to-cell communication. METHODS: A systematic examination of literature was conducted to gather relevant studies elucidating the biogenesis, composition, and functions of exosomes in the context of spermatogenesis. RESULTS: Exosomes play a pivotal role in orchestrating the complex signaling networks required for proper spermatogenesis. They facilitate the transfer of key regulatory molecules between different cell populations within the testes, including Sertoli cells, Leydig cells, and germ cells. CONCLUSION: The emerging understanding of exosome-mediated communication sheds light on novel mechanisms underlying spermatogenesis regulation. Further research in this area holds promise for insights into male reproductive health and potential therapeutic interventions.


Asunto(s)
Exosomas , Infertilidad Masculina , Espermatogénesis , Masculino , Espermatogénesis/fisiología , Exosomas/metabolismo , Humanos , Infertilidad Masculina/terapia , Infertilidad Masculina/metabolismo , Animales , Comunicación Celular , Células de Sertoli/metabolismo , Testículo/metabolismo , Células Intersticiales del Testículo/metabolismo , Transducción de Señal
2.
Biol Cell ; 116(4): e2300123, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38470182

RESUMEN

The testicular niche, which includes the germ cells, somatic cells, and extracellular matrix, plays a crucial role in maintaining the proper functions of the testis. Gonadotoxic treatments, such as chemotherapy and radiation therapy, have significantly improved the survival rates of cancer patients but have also been shown to have adverse effects on the testicular microenvironment. Therefore, repairing the testicular niche after gonadotoxic treatments is essential to restore its function. In recent years, several approaches, such as stem cell transplantation, gene therapy, growth factor therapy, and pharmacological interventions have been proposed as potential therapeutic strategies to repair the testicular niche. This comprehensive review aims to provide an overview of the current understanding of testis damage and repair mechanisms. We will cover a range of topics, including the mechanism of gonadotoxic action, repair mechanisms, and treatment approaches. Overall, this review highlights the importance of repairing the testicular niche after gonadotoxic treatments and identifies potential avenues for future research to improve the outcomes for cancer survivors.


Asunto(s)
Neoplasias , Testículo , Masculino , Humanos , Testículo/metabolismo , Neoplasias/terapia , Neoplasias/metabolismo , Microambiente Tumoral
3.
Zygote ; 32(1): 87-95, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38149356

RESUMEN

Mouse testicular tissue is composed of seminiferous tubules and interstitial tissue. Mammalian spermatogenesis is divided into three stages: spermatocytogenesis (mitotic divisions) in which spermatogonial stem cells (SSCs) turn into spermatocytes, followed by two consecutive meiotic divisions in which spermatocytes form spermatids. Spermatids differentiate into spermatozoa during spermiogenesis. Various factors affect the process of spermatogenesis and the organization of cells in the testis. Any disorder in different stages of spermatogenesis will have negative effects on male fertility. The aim of the current study was to compare the in vitro and in vivo spermatogenesis processes before and after transplantation to azoospermic mice using ultrastructural techniques. In this study, mice were irradiated with single doses of 14 Gy 60Co radiation. SSCs isolated from neonatal mice were cultured in vitro for 1 week and were injected into the seminiferous tubule recipient's mice. Testicular cells of neonatal mice were cultured in the four groups on extracellular matrix-based 3D printing scaffolds. The transplanted testes (8 weeks after transplantation) and cultured testicular cells in vitro (after 3 weeks) were then processed for transmission electron microscopy studies. Our study's findings revealed that the morphology and ultrastructure of testicular cells after transplantation and in vitro culture are similar to those of in vivo spermatogenesis, indicating that spermatogenic cell nature is unaltered in vitro.


Asunto(s)
Túbulos Seminíferos , Espermatogonias , Masculino , Ratones , Animales , Testículo , Espermatozoides , Espermatogénesis , Células Madre , Mamíferos
4.
Cell Tissue Res ; 394(3): 393-421, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37721632

RESUMEN

Men's reproductive health exclusively depends on the appropriate maturation of certain germ cells known as sperm. Certain illnesses, such as Klinefelter syndrome, cryptorchidism, and syndrome of androgen insensitivity or absence of testis maturation in men, resulting in the loss of germ cells and the removal of essential genes on the Y chromosome, can cause non-obstructive azoospermia. According to laboratory research, preserving, proliferating, differentiating, and transplanting spermatogonial stem cells or testicular tissue could be future methods for preserving the fertility of children with cancer and men with azoospermia. Therefore, new advances in stem cell research may lead to promising therapies for treating male infertility. The rate of progression and breakthrough in the area of in vitro spermatogenesis is lower than that of SSC transplantation, but newer methods are also being developed. In this regard, tissue and cell culture, supplements, and 3D scaffolds have opened new horizons in the differentiation of stem cells in vitro, which could improve the outcomes of male infertility. Various 3D methods have been developed to produce cellular aggregates and mimic the organization and function of the testis. The production of an artificial reproductive organ that supports SSCs differentiation will certainly be a main step in male infertility treatment.


Asunto(s)
Azoospermia , Infertilidad Masculina , Niño , Masculino , Humanos , Testículo , Espermatogonias , Semen , Espermatogénesis , Infertilidad Masculina/terapia
5.
Artif Organs ; 47(12): 1818-1830, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37698035

RESUMEN

PURPOSE: Development of organoids using human primary testicular cells has remained a challenge due to the complexity of the mammalian testicular cytoarchitecture and culture methods. In this study, we generated testicular organoids derived from human primary testicular cells. Then, we evaluated the effect of stem cell factor (SCF) on cell differentiation and apoptosis in the testicular organoid model. METHODS: The testicular cells were harvested from the three brain-dead donors. Human spermatogonial stem cells (SSCs) were characterized using immunocytochemistry (ICC), RT-PCR and flow cytometry. Testicular organoids were generated from primary testicular cells by hanging drop culture method and were cultured in three groups: control group, experimental group 1 (treated FSH and retinoic acid (RA)), and experimental group 2 (treated FSH, RA and SCF), for five weeks. We assessed the expression of SCP3 (Synaptonemal Complex Protein 3) as a meiotic gene, PRM2 (Protamine 2) as a post-meiotic marker and apoptotic genes of Bax (BCL2-Associated X Protein) and Bcl-2 (B-cell lymphoma 2), respectively by using RT-qPCR. In addition, we identified the expression of PRM2 by immunohistochemistry (IHC). RESULTS: Relative expression of SCP3, PRM2 and Bcl-2 were highest in group 2 after five weeks of culture. In contrast, BAX expression level was lower in experimental group 2 in comparison with other groups. IHC analyses indicated the highest expression of PRM2 as a postmeiotic marker in group 2 in comparison to 2D culture and control groups but not find significant differences between experimental group 1 and experimental group 2 groups. Morphological evaluations revealed that organoids are compact spherical structures and in the peripheral region composed of uncharacterized elongated fibroblast-like cells. CONCLUSION: Our findings revealed that the testicular organoid culture system promote the spermatogonial stem cell (SSC) differentiation, especially in presence of SCF. Developed organoids are capable of recapitulating many important properties of a stem cell niche.


Asunto(s)
Espermatogonias , Factor de Células Madre , Masculino , Animales , Humanos , Factor de Células Madre/farmacología , Factor de Células Madre/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/farmacología , Espermatogonias/metabolismo , Espermatogénesis/genética , Diferenciación Celular , Organoides , Hormona Folículo Estimulante/metabolismo , Hormona Folículo Estimulante/farmacología , Células Cultivadas , Mamíferos
6.
J Assist Reprod Genet ; 39(1): 153-163, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34519944

RESUMEN

PURPOSE: The generation of germ cells from mesenchymal stromal cells (MSCs) provides a valuable in vitro platform for infertility modeling. The establishment of these cells is a new approach for assisted reproductive technology (ART) to help infertile patients who lack functional gametes. METHODS: Human adipose-derived MSCs were isolated and then characterized for multipotency by flow cytometry, differentiation capacity, and cytogenetic assays. These cells were used in a male germ cell differentiation study. The expression of male germ cell markers was evaluated at day 21 of differentiation using an immunofluorescence assay, flow cytometry, and RT-qPCR. Undifferentiated MSCs were used for transplantation in busulfan-induced azoospermic mice. RESULTS: In this study, MSCs were successfully isolated from human adipose tissues which were positive for cell markers such as CD90, CD105, CD73, and CD29 but negative for CD34 and CD45. The results of flow cytometry, immunocytochemistry, and RT-qPCR analysis at day 21 of differentiation showed that the undifferentiated adipose-derived MSCs are able to differentiate into male germ cells. Additionally, transplantation of undifferentiated MSCs in busulfan-induced azoospermic mice caused spermatogenesis recovery in the majority of seminiferous tubules. CONCLUSION: In this study, we showed that differentiation of human adipose-derived MSCs into male germ cells is a useful tool for in vitro study of human germ cell development. Our results demonstrated that cell therapy with adipose-derived MSCs could help the repair of pathological changes in testicular seminiferous tubules. Therefore, it may have a clinical application for the treatment of azoospermia in infertile patients.


Asunto(s)
Azoospermia/tratamiento farmacológico , Células Madre Mesenquimatosas/metabolismo , Animales , Azoospermia/etiología , Azoospermia/fisiopatología , Busulfano/efectos adversos , Modelos Animales de Enfermedad , Inmunosupresores/efectos adversos , Masculino , Células Madre Mesenquimatosas/inmunología , Ratones , Espermatogénesis/efectos de los fármacos , Espermatogénesis/genética
7.
J Mater Sci Mater Med ; 32(5): 47, 2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33891169

RESUMEN

Decellularized scaffolds have been found to be excellent platforms for tissue engineering applications. The attempts are still being made to optimize a decellularization protocol with successful removal of the cells with minimal damages to extracellular matrix components. We examined twelve decellularization procedures using different concentrations of Sodium dodecyl sulfate and Triton X-100 (alone or in combination), and incubation time points of 15 or 30 min. Then, the potential of the decellularized scaffold as a three-dimensional substrate for colony formation capacity of mouse spermatogonial stem cells was determined. The morphological, degradation, biocompatibility, and swelling properties of the samples were fully characterized. The 0.5%/30 SDS/Triton showed optimal decellularization with minimal negative effects on ECM (P ≤ 0.05). The swelling ratios increased with the increase of SDS and Triton concentration and incubation time. Only 0.5%/15 and 30 SDS showed a significant decrease in the SSCs viability compared with other groups (P < 0.05). The SSCs colony formation was clearly observed under SEM and H&E stained slides. The cells infiltrated into the subcutaneously implanted scaffold at days 7 and 30 post-implantation with no sign of graft rejection. Our data suggest the %0.5/30 SDS/Triton as an excellent platform for tissue engineering and reproductive biology applications.


Asunto(s)
Células Madre Germinales Adultas/fisiología , Movimiento Celular/fisiología , Matriz Extracelular/química , Placenta/efectos de los fármacos , Andamios del Tejido , Animales , Animales Recién Nacidos , Femenino , Humanos , Ratones , Octoxinol/química , Embarazo , Dodecil Sulfato de Sodio/química , Ingeniería de Tejidos/métodos
8.
Andrologia ; 53(1): e13799, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33099803

RESUMEN

The molecular mechanisms of drug use on sexual health are largely unknown. We investigated, the relationship between heroin use disorder and epigenetic factors influencing histone acetylation in sperm cells. The volunteers included twenty-four 20- to 50-year-old men with a normal spermogram who did not consume any drugs and twenty-four age- to BMI-matched men who consume only the drug heroin for more than last four months. HDAC1 and HDAC11 mRNA expression levels in spermatozoa and miR-34c-5p and miR-125b-5p expression levels in seminal plasma were measured. The heroin-user group showed significantly increased white blood cell counts and decreased sperm motility and survival rates (8.61 ± 1.73, 21.50 ± 3.11, 69.90 ± 4.69 respectively) as compared to the control group (1.49 ± 0.32, 38.82 ± 3.05, 87.50 ± 0.99 respectively) (p ≤ .001). An increase in DNA fragmentation index (DFI) (heroin-user group: 41.93 ± 6.59% and control group: 10.14 ± 1.43%, p = .003), a change in frequency of HDAC1 (heroin-user group: 1.69 ± 0.55 and control group: 0.45 ± 0.14, p = .045) and HDAC11 (heroin-user group: 0.29 ± 0.13 and control group: 2.36 ± 0.76, p = .019) in spermatozoa and a significant decrease in seminal miR-125b-5p abundance (heroin-user group: 0.37 ± 0.11 and control group: 1.59 ± 0.47, p = .028) were reported in heroin consumers. Heroin use can lead to male infertility by causing leukocytospermia, asthenozoospermia, DFI elevation in sperm cells and alterations in seminal RNA profile.


Asunto(s)
Heroína , Infertilidad Masculina , Adulto , Fragmentación del ADN , Epigénesis Genética , Heroína/toxicidad , Histona Desacetilasas , Humanos , Infertilidad Masculina/genética , Masculino , Persona de Mediana Edad , Semen , Motilidad Espermática , Espermatozoides , Adulto Joven
9.
Med J Islam Repub Iran ; 35: 129, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35321367

RESUMEN

Background: A wide variety of cytokines are released from human amniotic membrane cells (hAMCs), which can increase the rate of differentiation of mesenchymal stem cells into the neurons. We studied the effect of Retinoic Acid (RA) on the differentiation rate of human Umbilical Cord Mesenchymal Stem Cells (hUMSCs) which were co-cultured with hAMCs. Methods: In this experimental study, both hUMSCs and hAMCs were isolated from postpartum human umbilical cords and placenta respectively. The expression of mesenchymal (CD73, CD90 and CD105), hematopoietic and endothelial (CD34 and CD45) markers in hUMSCs were confirmed by flow cytometry. The hUMSCs were cultured in four distinct groups: group 1) Control, group 2) Co-culture with hAMCs, group 3) RA treatment and group 4) Co-culture with hAMCs treated by RA. Twelve days after culturing, the expression of NSE, MAP2 and ChAT differentiation genes and their related proteins were examined by real-time PCR and immunocytochemistry respectively. Results: The flow-cytometry analysis indicated increased expression of mesenchymal markers and a low expression of both hematopoietic and endothelial markers (CD73:98.24%, CD90: 97.32%, CD105: 90.75%, CD34: 2.96%, and CD45:1.74%). Moreover, the expression of both NSE and MAP2 markers was increased significantly in all studied groups in comparison to the control group On the other hand, the expression of ChAT had a significant increase in the group 2 and 4 (RA and RA+ co-culture). Conclusion: RA can be used as an effective inducer to differentiate hUMSCs into cholinergic-like cells, and hAMCs could increase the number of differentiated cells as an effective factor.

10.
Exp Cell Res ; 384(1): 111544, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31479684

RESUMEN

Cells grow differently in conventional 2D cell culture than when they grow in the physiological microenvironment. In this study, we developed a 3D cell culture model for generating male germ cells from human iPSCs using a human decellularized amnion membrane (DAM) scaffold. To this end, human iPSCs were generated using retroviral vectors and characterized for pluripotency properties by immunofluorescence assay, flow cytometry, ALP staining, cytogenetic assay, and differentiation capacity. The iPSCs were used for investigating male germ cells differentiation efficiency in both conventional 2D culture and 3D-DAM scaffold. The expression of male germ cell markers was evaluated at day 21 of differentiation using immunofluorescence assay, flow-cytometry, and RT-qPCR. The results indicated a successful reprogramming of human foreskin fibroblast cells into pluripotent iPSCs. The reprogrammed cells were positive for pluripotency markers and differentiated into the three germ layers. During male germ cell differentiation, the cells tend to aggregate and form colony-like structures in both 2D and 3D conditions. However, significant expression of VASA, DAZL, PLZF, STELLA, and NANOS3 markers and more efficient haploid male germ cell production were observed in the 3D condition when compared to the 2D model. Considering the effect of the 3D-DAM scaffold in prompting male germ cell-specific markers and increased efficiency of germ cell differentiation in 3D culture, it appears that DAM scaffold is a useful tool for in vitro studies of human germ cell development and ultimately future clinical application.


Asunto(s)
Amnios/citología , Células Madre Pluripotentes Inducidas/citología , Amnios/metabolismo , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Células Germinativas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Andamios del Tejido
11.
J Cell Biochem ; 120(8): 12508-12518, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30977186

RESUMEN

In this study, we present an electrospun gelatin (EG) scaffold to mimic the extracellular matrix of the testis. The EG scaffold was synthesized by electrospinning and crosslinked with glutaraldehyde vapor to decrease its water solubility and degradation rate. The scanning electron microscope micrographs showed the homogenous morphology of randomly aligned gelatin fibers. The average diameter of gelatin fibers before and after crosslinking was approximately 180 and 220 nm, respectively. Modulus, tensile strength, and elongation at break values were as 161.8 ± 24.4 MPa, 4.21 ± 0.54 MPa, and 7.06 ± 2.12 MPa, respectively. The crosslinked EG showed 75.2% ± 4.5% weight loss after 14 days with no changes in the pH value of degradation solution. Cytobiocompatibility of the EG for sertoli cells and embryonic stem cells (ESCs) was determined in vitro. Sertoli cells were isolated from mouse testis and characterized by immunostaining and flow cytometry. The effects of EG on proliferation and attachment of both sertoli cells and ESCs were examined. The EG scaffolds exhibited no cytotoxicity for sertoli and ESCs. Both sertoli and ESCs were well attached and grown on EG. Coculture of sertoli and ESCs on EG showed better ESCs adhesion compared with ESCs alone. Our findings indicate the potential of EG as a substrate for proliferation, adhesion, and coculture of sertoli and ESCs and may be considered as a promising engineered microenvironment for in vitro coculture system with the aim of guiding stem cells differentiation toward sperm-producing cells.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Madre Embrionarias/fisiología , Gelatina , Células de Sertoli/fisiología , Andamios del Tejido , Animales , Proliferación Celular , Matriz Extracelular , Masculino , Ratones , Testículo
12.
Clin Sci (Lond) ; 131(18): 2381-2395, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28768794

RESUMEN

The present study aimed to: (i) identify the exogenous factors that allow in vitro differentiation of mouse spermatogonial stem cells (SSCs) from embryonic stem cells (ESCs); (ii) evaluate the effects of Sertoli cells in SSC enrichment; and (iii) assess the success of transplantation using in vitro differentiated SSCs in a mouse busulfan-treated azoospermia model. A 1-day-old embryoid body (EB) received 5 ng/ml of bone morphogenetic protein 4 (BMP4) for 4 days, 3 µM retinoic acid (RA) in a SIM mouse embryo-derived thioguanine and ouabain resistant (STO) co-culture system for 7 days, and was subsequently co-cultured for 2 days with Sertoli cells in the presence or absence of a leukaemia inhibitory factor (LIF), basic fibroblast growth factor (bFGF) and RA composition, and in the presence of these factors in simple culture medium. Higher viability, proliferation and germ cell gene expression were seen in the presence of the LIF, bFGF and RA composition, on top of Sertoli cells. Immunocytochemistry results showed higher CDH1 expression in this group. Sertoli co-culture had no effects on SSC proliferation. Eight weeks after transplantation, injected cells were observed at the base of the seminiferous tubules and in the recipient testes. The number of spermatogonia and the mass of the testes were higher in transplanted testes relative to the control group. It seems that transplantation of these cells can be useful in infertility treatment.


Asunto(s)
Azoospermia/cirugía , Células Madre Embrionarias/fisiología , Espermatogénesis/fisiología , Espermatozoides/trasplante , Animales , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Células de Sertoli/fisiología , Testículo/cirugía
13.
Arch Gynecol Obstet ; 295(4): 1041-1050, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28271235

RESUMEN

PURPOSE: AKTs have a pivotal role in the granulosa-lutein cell (GC) proliferation and folliculogenesis, and there is a reciprocal feedback between AKT with androgen. Therefore, we aimed to evaluate the role of AKTs in GCs of hyperandrogenic (+HA) PCOS cases. METHOD: There were three groups: control, +HA PCOS and -HA (non-hyperandrogenic) PCOS. All groups were subjected to GnRH antagonist protocol for stimulation of ovulation. Follicular fluid was aspirated from large follicles, and GCs were isolated using cell strainer method. AKT1, AKT2, AKT3, and androgen receptor (AR) mRNA expressions were analyzed with quantitative real-time PCR (qRT-PCR), and total-AKT and p-AKT (Ser473 & Thr308) were investigated using western blotting. RESULTS: There were high levels of AKT1, AKT2, and AR mRNA expressions and high levels of p-AKT protein expression in the +HA PCOS group (p ≤ 0.05). There was a direct positive correlation between free testosterone (FT) and total testosterone (TT) with the levels of AKT1, AKT2, and p-AKT (Ser473), and also between FT with the levels of AR. CONCLUSION: High expressions of AKT1 and AKT2 through possible relation with androgen may cause GCs dysfunction in the +HA PCOS patients.


Asunto(s)
Células de la Granulosa/metabolismo , Hiperandrogenismo/complicaciones , Células Lúteas/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adulto , Femenino , Líquido Folicular/metabolismo , Humanos , Hiperandrogenismo/metabolismo , Modelos Lineales , Inducción de la Ovulación , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/fisiopatología , Proteínas Proto-Oncogénicas c-akt/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Testosterona/sangre
14.
J Assist Reprod Genet ; 33(6): 771-81, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27052833

RESUMEN

PURPOSE: Testicular ischemia is the main consequence of testicular torsion, in both clinical and experimental aspects. Preservation and auto-transplantation of spermatogonial stem cells (SSCs) could be a new treatment for infertility in testicular ischemia following testicular torsion. METHODS: To apply the idea in this study, animals were randomly divided into four groups of control, sham, with torsion, and with torsion followed by transplantation (TT). Isolated SSCs from neonatal mice were cultured and identified by flow cytometry (C-KIT(-), INTEGRIN ß1 (+)) and RT-PCR (Reverse transcription polymerase chain reaction) for specific spermatogonial cell markers (Oct4, Gfrα-1, Plzf, Vasa, Itgα 6 , and Itgß 1 ). SSCs were transplanted upon a 2-h testicular torsion in the TT group. Cultured cells were transplanted into ischemia reperfusion testicle 2 weeks post-testicular torsion. Eight weeks after SSCs transplantation, the SSCs-transplanted testes and epididymis were removed for sperm analysis, weight and histopathological evaluation, and pre- and post-meiotic gene expression assessment by qRT-PCR. RESULTS: Our findings indicated that all evaluated parameters (epididymal sperm profile, Johnsen score, Plzf, Gfrα-1, Scp-1, Tekt-1 expressions, and histopathological profile) were significantly decreased following testicular torsion (group 3) when compared to the control group (p ≤ 0.05). However, all abovementioned parameters showed a significant increase/improvement in torsion-transplantation group compared to torsion group. However, these parameters in the TT group were significantly lower in the sham and control groups (p ≤ 0.05). CONCLUSION: SSCs transplantation could up-regulate the expression of pre- and post-meiotic genes in testicular ischemia, which resulted in improvement of both testicular function and structure after testicular torsion.


Asunto(s)
Torsión del Cordón Espermático/patología , Espermatogénesis , Espermatogonias/trasplante , Animales , Biomarcadores/metabolismo , Perfilación de la Expresión Génica , Masculino , Ratones , Espermatogonias/metabolismo , Trasplante de Células Madre
15.
Clin Exp Reprod Med ; 51(3): 171-180, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38525520

RESUMEN

Male infertility can be caused by genetic anomalies, endocrine disorders, inflammation, and exposure to toxic chemicals or gonadotoxic treatments. Therefore, several recent studies have concentrated on the preservation and restoration of fertility to enhance the quality of life for affected individuals. It is currently recommended to biobank the tissue extracted from testicular biopsies to provide a later source of spermatogonial stem cells (SSCs). Another successful approach has been the in vitro production of haploid male germ cells. The capacity of SSCs to transform into sperm, as in testicular tissue transplantation, SSC therapy, and in vitro or ex vivo spermatogenesis, makes them ideal candidates for in vivo fertility restoration. The transplantation of SSCs or testicular tissue to regenerate spermatogenesis and create embryos has been achieved in nonhuman mammal species. Although the outcomes of human trials have yet to be released, this method may soon be approved for clinical use in humans. Furthermore, regenerative medicine techniques that develop tissue or cells on organic or synthetic scaffolds enriched with bioactive molecules have also gained traction. All of these methods are now in different stages of experimentation and clinical trials. However, thanks to rigorous studies on the safety and effectiveness of SSC-based reproductive treatments, some of these techniques may be clinically available in upcoming decades.

16.
Macromol Biosci ; 24(2): e2300342, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37729950

RESUMEN

Numerous scaffolds are developed in the field of testicular bioengineering. However, effectively replicating the spatial characteristics of native tissue, poses a challenge in maintaining the requisite cellular arrangement essential for spermatogenesis. In order to mimic the structural properties of seminiferous tubules, the objective is to fabricate a biocompatible tubular scaffold. Following the decellularization process of the testicular tissue, validation of cellular remnants' elimination from the specimens is conducted using 4',6-diamidino-2-phenylindole staining, hematoxylin and eosin staining, and DNA content analysis. The presence of extracellular matrix (ECM) components is confirmed through Alcian blue, Orcein, and Masson's trichrome staining techniques. The electrospinning technique is employed to synthesize the scaffolds using polycaprolactone (PCL), extracted ECM, and varying concentrations of graphene oxide (GO) (0.5%, 1%, and 2%). Subsequently, comprehensive evaluations are performed to assess the properties of the synthetic scaffolds. These evaluations encompass Fourier-transform infrared spectroscopy, scanning electron microscopy imaging, scaffold degradation testing, mechanical behavior analysis, methylthiazolyldiphenyl-tetrazolium bromide assay, and in vivo biocompatibility assessment. The PCL/decellularized extracellular matrix with 0.5% GO formulation exhibits superior fiber morphology and enhanced mechanical properties, and outperforms other groups in terms of in vitro biocompatibility. Consequently, these scaffolds present a viable option for implementation in "in vitro spermatogenesis" procedures, holding promise for future sperm production from spermatogonial cells.


Asunto(s)
Grafito , Medicina Reproductiva , Andamios del Tejido , Masculino , Humanos , Andamios del Tejido/química , Ingeniería de Tejidos/métodos , Biomimética , Semen , Poliésteres/farmacología , Poliésteres/química , Matriz Extracelular/química , Túbulos Seminíferos
17.
J Biomed Mater Res B Appl Biomater ; 112(5): e35414, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38733611

RESUMEN

Utilizing natural scaffold production derived from extracellular matrix components presents a promising strategy for advancing in vitro spermatogenesis. In this study, we employed decellularized human placental tissue as a scaffold, upon which neonatal mouse spermatogonial cells (SCs) were cultured three-dimensional (3D) configuration. To assess cellular proliferation, we examined the expression of key markers (Id4 and Gfrα1) at both 1 and 14 days into the culture. Our quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis revealed a notable increase in Gfrα1 gene expression, with the 3D culture group exhibiting the highest levels. Furthermore, the relative frequency of Gfrα1-positive cells significantly rose from 38.1% in isolated SCs to 46.13% and 76.93% in the two-dimensional (2D) and 3D culture systems, respectively. Moving forward to days 14 and 35 of the culture period, we evaluated the expression of differentiating markers (Sycp3, acrosin, and Protamine 1). Sycp3 and Prm1 gene expression levels were upregulated in both 2D and 3D cultures, with the 3D group displaying the highest expression. Additionally, acrosin gene expression increased notably within the 3D culture. Notably, at the 35-day mark, the percentage of Prm1-positive cells in the 3D group (36.4%) significantly surpassed that in the 2D group (10.96%). This study suggests that the utilization of placental scaffolds holds significant promise as a bio-scaffold for enhancing mouse in vitro spermatogenesis.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Placenta , Animales , Femenino , Ratones , Masculino , Humanos , Placenta/citología , Placenta/metabolismo , Embarazo , Espermatogonias/citología , Espermatogonias/metabolismo , Andamios del Tejido/química , Matriz Extracelular Descelularizada/química , Matriz Extracelular Descelularizada/metabolismo , Células Madre/metabolismo , Células Madre/citología
18.
Clin Exp Reprod Med ; 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38853126

RESUMEN

Among the primary objectives of contemporary assisted reproductive technology research are achieving the births of healthy singletons and improving overall fertility outcomes. Substantial advances have been made in refining the selection of single embryos for transfer, with the aim of maximizing the likelihood of successful implantation. The principal criterion for this selection is embryo morphology. Morphological evaluation systems are based on traditional parameters, including cell count and fragmentation, pronuclear morphology, cleavage rate, blastocyst formation, and various sequential embryonic assessments. To reduce the incidence of multiple pregnancies and to identify the single embryo with the highest potential for growth, invasive techniques such as preimplantation genetic screening are employed in in vitro fertilization clinics. However, new approaches have been suggested for clinical application that do not harm the embryo and that provide consistent, accurate results. Noninvasive technologies, such as time-lapse imaging and omics, leverage morphokinetic parameters and the byproducts of embryo metabolism, respectively, to identify noninvasive prognostic markers for competent single embryo selection. While these technologies have garnered considerable interest in the research community, they are not incorporated into routine clinical practice and still have substantial room for improvement. Currently, the most promising strategies involve integrating multiple methodologies, which together are anticipated to increase the likelihood of successful pregnancy.

19.
Int J Biol Macromol ; 277(Pt 4): 134362, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39089552

RESUMEN

Healing diabetic ulcers with chronic inflammation is a major challenge for researchers and professionals, necessitating new strategies. To rapidly treat diabetic wounds in rat models, we have fabricated a composite scaffold composed of alginate (Alg) and silk fibroin (SF) as a wound dressing that is laden with molecules of lithium chloride (LC). The physicochemical, bioactivity, and biocompatibility properties of Alg-SF-LC scaffolds were investigated in contrast to those of Alg, SF, and Alg-SF ones. Afterward, full-thickness wounds were ulcerated in diabetic rats in order to evaluate the capacity of LC-laden scaffolds to regenerate skin. The characterization findings demonstrated that the composite scaffolds possessed favorable antibacterial properties, cell compatibility, high swelling, controlled degradability, and good uniformity in the interconnected pore microstructure. Additionally, in terms of wound contraction, re-epithelialization, and angiogenesis improvement, LC-laden scaffolds revealed better performance in diabetic wound healing than the other groups. This research indicates that utilizing lithium chloride molecules loaded in biological materials supports the best diabetic ulcer regeneration in vivo, and produces a skin replacement with a cellular structure comparable to native skin.


Asunto(s)
Alginatos , Antibacterianos , Diabetes Mellitus Experimental , Fibroínas , Cloruro de Litio , Andamios del Tejido , Cicatrización de Heridas , Fibroínas/química , Fibroínas/farmacología , Animales , Cicatrización de Heridas/efectos de los fármacos , Alginatos/química , Alginatos/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Ratas , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/complicaciones , Porosidad , Andamios del Tejido/química , Cloruro de Litio/farmacología , Cloruro de Litio/química , Neovascularización Fisiológica/efectos de los fármacos , Masculino , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología
20.
J Assist Reprod Genet ; 30(3): 325-32, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23274510

RESUMEN

PURPOSE: Spermatogonial stem cells are affected by the interactions of extrinsic signals produced by components of the microenvironment niche, in addition to the chemical and physical properties of the extracellular matrix. Therefore, this study was initiated to assess the interaction of these cells on a synthetic nanofibrillar extracellular matrix that mimicked the geometry and nanotopography of the basement membrane for cellular growth. METHODS: This study has used a variety of experimental approaches to investigate the interaction of mouse neonatal-derived spermatogonial stem-like cells on a synthetic random oriented three-dimensional nanofibrillar matrix composed of electrospun polyamide nanofibers (Ultra-Web™). RESULTS: Spermatogonial stem-like cell colonies were characterized by their ability to express α6-integrin, Thy-1, PLZF, and ß1-integrin. After culture of cells on the nanofibrillar surfaces for 7 days, the number of colonies, the number of cells in each colony, and the average area of colonies were increased (P < 0.05). However, the expression difference of related markers in both groups was not significant. A significantly higher proliferation and survival was observed in the nanofibrillar group (P < 0.05). After transplantation into the testes of busulfan-treated adult mice, spermatogonial stem-like cell colonies that were cultured on the nanofibrillar surface demonstrated functionality, as verified by their ability to migrate to the seminiferous basal membrane, where they produced additional colonies. CONCLUSIONS: These results have suggested that electrospun nanofibrillar surfaces could provide a more favorable microenvironment for in vitro short term culture of spermatogonial stem-like cell colonies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Nanofibras/química , Espermatogonias/citología , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Femenino , Masculino , Ratones , Embarazo , Células Madre/metabolismo , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA