Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nanoscale Horiz ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38952214

RESUMEN

Zinc-ion batteries (ZIBs) are regarded as promising power sources for flexible and biocompatible devices due to their good sustainability and high intrinsic safety. However, their applications have been hindered by the issues of uncontrolled Zn dendrite growth and severe water-induced side reactions in conventional liquid electrolytes. Herein, an ionically cross-linked composite hydrogel electrolyte based on natural biomacromolecules, including iota-carrageenan and sodium alginate, is designed to promote highly efficient and reversible Zn plating/stripping. The abundant functional groups of macromolecules effectively suppress the reactivity of water molecules and facilitate uniform Zn deposition. Moreover, the composite hydrogel electrolyte exhibits a high ionic conductivity of 5.89 × 10-2 S cm-1 and a Zn2+ transference number of 0.58. Consequently, the Zn‖Zn symmetric cell with the composite hydrogel electrolyte shows a stable cycle life of more than 500 h. Meanwhile, the Zn‖NH4V4O10 coin cell with the composite hydrogel electrolyte retains a high specific capacity of approximately 200 mA h g-1 after 600 cycles at 2 A g-1. The Zn‖NVO pouch cell based on the composite hydrogel electrolyte also shows a high specific capacity of 246.1 mA h g-1 at 0.5 A g-1 and retains 70.7% of its initial capacity after 150 cycles. The pouch cell performs well at different bending angles and exhibits a capacity retention rate of 98% after returning to its initial state from 180° folding. This work aims to construct high-performance hydrogel electrolytes using low-cost natural materials, which may provide a solution for the application of ZIBs in flexible biocompatible devices.

2.
Cell Mol Life Sci ; 81(1): 108, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38421455

RESUMEN

Spermiogenesis is considered to be crucial for the production of haploid spermatozoa with normal morphology, structure and function, but the mechanisms underlying this process remain largely unclear. Here, we demonstrate that SPEM family member 2 (Spem2), as a novel testis-enriched gene, is essential for spermiogenesis and male fertility. Spem2 is predominantly expressed in the haploid male germ cells and is highly conserved across mammals. Mice deficient for Spem2 develop male infertility associated with spermiogenesis impairment. Specifically, the insufficient sperm individualization, failure of excess cytoplasm shedding, and defects in acrosome formation are evident in Spem2-null sperm. Sperm counts and motility are also significantly reduced compared to controls. In vivo fertilization assays have shown that Spem2-null sperm are unable to fertilize oocytes, possibly due to their impaired ability to migrate from the uterus into the oviduct. However, the infertility of Spem2-/- males cannot be rescued by in vitro fertilization, suggesting that defective sperm-egg interaction may also be a contributing factor. Furthermore, SPEM2 is detected to interact with ZPBP, PRSS21, PRSS54, PRSS55, ADAM2 and ADAM3 and is also required for their processing and maturation in epididymal sperm. Our findings establish SPEM2 as an essential regulator of spermiogenesis and fertilization in mice, possibly in mammals including humans. Understanding the molecular role of SPEM2 could provide new insights into future therapeutic treatment of human male infertility and development of non-hormonal male contraceptives.


Asunto(s)
Infertilidad Masculina , Testículo , Humanos , Femenino , Masculino , Animales , Ratones , Semen , Espermatogénesis/genética , Infertilidad Masculina/genética , Interacciones Espermatozoide-Óvulo , Mamíferos , Fertilinas
3.
Reprod Sci ; 30(1): 145-168, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35471551

RESUMEN

Our previous studies have reported that a putative trypsin-like serine protease, PRSS37, is exclusively expressed in testicular germ cells during late spermatogenesis and essential for sperm migration from the uterus into the oviduct and sperm-egg recognition via mediating the interaction between PDILT and ADAM3. In the present study, the global proteome profiles of wild-type (wt) and Prss37-/- mice in testis and sperm were compared employing data independent acquisition (DIA) technology. Overall, 2506 and 459 differentially expressed proteins (DEPs) were identified in Prss37-null testis and sperm, respectively, when compared to control groups. Bioinformatic analyses revealed that most of DEPs were related to energy metabolism. Of note, the DEPs associated with pathways for the catabolism such as glucose via glycolysis, fatty acids via ß-oxidation, and amino acids via oxidative deamination were significantly down-regulated. Meanwhile, the DEPs involved in the tricarboxylic acid cycle (TCA cycle) and oxidative phosphorylation (OXPHOS) were remarkably decreased. The DIA data were further confirmed by a markedly reduction of intermediate metabolites (citrate and fumarate) in TCA cycle and terminal metabolite (ATP) in OXPHOS system after disruption of PRSS37. These outcomes not only provide a more comprehensive understanding of the male fertility of energy metabolism modulated by PRSS37 but also furnish a dynamic proteomic resource for further reproductive biology studies.


Asunto(s)
Proteómica , Serina Proteasas , Testículo , Animales , Femenino , Masculino , Ratones , Metabolismo Energético , Proteína Disulfuro Isomerasas/metabolismo , Semen/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Serina Proteasas/deficiencia , Serina Proteasas/genética , Ratones Noqueados
4.
Biol Reprod ; 107(4): 1139-1154, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-35863763

RESUMEN

Serine proteases (PRSS) constitute nearly one-third of all proteases, and many of them have been identified to be testis-specific and play significant roles during sperm development and male reproduction. PRSS54 is one of the testis-specific PRSS in mouse and human but its physiological function remains largely unclear. In the present study, we demonstrate in detail that PRSS54 exists not only in testis but also in mature sperm, exhibiting a change in protein size from 50 kDa in testis to 42 kDa in sperm. Loss of PRSS54 in mice results in male subfertility, acrosome deformation, defective sperm-zona penetration, and phenotypes of male subfertility and acrosome deformation can be rescued by Prss54 transgene. Ultrastructure analyses by transmission electronic microscopy further reveal various morphological abnormalities of Prss54-/- spermatids during spermiogenesis, including unfused vacuoles in acrosome, detachment and eccentrical localization of the acrosomal granules, and asymmetrical elongation of the nucleus. Subcellular localization of PRSS54 display that it appears in the acrosomal granule at the early phase of acrosome biogenesis, then extends along the inner acrosomal membrane, and ultimately presents in the acrosome region of the mature sperm. PRSS54 interacts with acrosomal proteins ZPBP1, ZPBP2, ACRBP, and ZP3R, and loss of PRSS54 affects the distribution of these proteins in testis and sperm, although their protein levels are largely unaffected. Moreover, Prss54-/- sperm are more sensitive to acrosome reaction inducers.


Asunto(s)
Acrosoma , Infertilidad Masculina , Acrosoma/metabolismo , Animales , Proteínas Portadoras/metabolismo , Proteínas del Huevo , Humanos , Infertilidad Masculina/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Morfogénesis , Proteínas/metabolismo , Semen/metabolismo , Serina Endopeptidasas/metabolismo , Serina Proteasas/genética , Serina Proteasas/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo
5.
J Cell Sci ; 134(10)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-34028541

RESUMEN

A disintegrin and metalloproteinase 3 (ADAM3) is a sperm membrane protein critical for sperm migration from the uterus into the oviduct and sperm-egg binding in mice. Disruption of PRSS37 results in male infertility concurrent with the absence of mature ADAM3 from cauda epididymal sperm. However, how PRSS37 modulates ADAM3 maturation remains largely unclear. Here, we determine the PRSS37 interactome by GFP immunoprecipitation coupled with mass spectrometry in PRSS37-EGFP knock-in mice. Three molecular chaperones (CLGN, CALR3 and PDILT) and three ADAM proteins (ADAM2, ADAM6B and ADAM4) were identified to be interacting with PRSS37. Coincidently, five of them (except ADAM4) have been reported to interact with ADAM3 precursor and regulate its maturation. We further demonstrated that PRSS37 also interacts directly with ADAM3 precursor and its deficiency impedes the association between PDILT and ADAM3. This could contribute to improper translocation of ADAM3 to the germ cell surface, leading to ADAM3 loss in PRSS37-null mature sperm. The understanding of the maturation mechanisms of pivotal sperm plasma membrane proteins will pave the way toward novel strategies for contraception and the treatment of unexplained male infertility.


Asunto(s)
Infertilidad Masculina , Glicoproteínas de Membrana , Proteínas ADAM/genética , Animales , Epidídimo , Femenino , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Proteína Disulfuro Isomerasas , Serina Proteasas , Espermatozoides
6.
J Bone Miner Res ; 36(4): 779-791, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33316109

RESUMEN

Fibroblast growth factor 9 (Fgf9) is a well-known factor that regulates bone development; however, its function in bone homeostasis is still unknown. Previously, we identified a point mutation in the FGF9 gene (p.Ser99Asn, S99N) and generated an isogeneic knock-in mouse model, which revealed that this loss-of-function mutation impaired early joint formation and was responsible for human multiple synostosis syndrome 3 (SYNS3). Moreover, newborn and adult S99N mutant mice exhibited significantly increased bone mass, suggesting that Fgf9 also participated in bone homeostasis. Histomorphology, tomography, and serological analysis of homozygous newborns and heterozygous adults showed that the Fgf9S99N mutation immensely increased bone mass and bone formation in perinatal and adult bones and decreased osteoclastogenesis in adult bone. An in vitro differentiation assay further revealed that the S99N mutation enhanced bone formation by promoting osteogenesis and mineralization of bone marrow mesenchymal stem cells (BMSCs) and attenuating osteoclastogenesis of bone marrow monocytes (BMMs). Considering the loss-of-function effect of the S99N mutation, we hypothesized that Fgf9 itself inhibits osteogenesis and promotes osteoclastogenesis. An in vitro differentiation assay revealed that Fgf9 prominently inhibited BMSC osteogenic differentiation and mineralization and showed for the first time that Fgf9 promoted osteoclastogenesis by enhancing preosteoclast aggregation and cell-cell fusion. Furthermore, specific inhibitors and in vitro differentiation assays were used and showed that Fgf9 inhibited BMSC osteogenesis mainly via the MEK/ERK pathway and partially via the PI3K/AKT pathway. Fgf9 also promoted osteoclastogenesis as a potential costimulatory factor with macrophage colony-stimating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) by coactivating the MAPK and PI3K/AKT signaling pathways. Taken together, our study demonstrated that Fgf9 is a negative regulator of bone homeostasis by regulating osteogenesis and osteoclastogenesis and provides a potential therapeutic target for bone degenerative diseases. © 2020 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Osteogénesis , Proteínas Proto-Oncogénicas c-akt , Adulto , Animales , Diferenciación Celular , Factor 9 de Crecimiento de Fibroblastos , Humanos , Recién Nacido , Ratones , FN-kappa B , Osteoclastos , Fosfatidilinositol 3-Quinasas , Ligando RANK
7.
J Reprod Dev ; 66(1): 57-65, 2020 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-31801914

RESUMEN

Testis-specific genes are prone to affect spermatogenesis or sperm fertility, and thus may play pivotal roles in male reproduction. However, whether a gene really affects male reproduction in vivo needs to be confirmed using a gene knock-out (KO) model, a 'gold standard' method. Increasing studies have found that some of the evolutionarily conserved testis-enriched genes are not essential for male fertility. In this study, we report that 1700121C10Rik, a previously uncharacterized gene, is specifically expressed in the testis and produces two long noncoding RNAs (lncRNAs) in mouse: Transcript 1 and Transcript 2. qRT-PCR, northern blotting, and in situ hybridization revealed that expression of both the lncRNAs commenced at the onset of sexual maturity and was predominant in round and elongating spermatids during spermiogenesis. Moreover, we found different subcellular localization of Transcript 1 and Transcript 2 that was predominant in the cytoplasm and nucleus, respectively. 1700121C10Rik-KO mouse model disrupting Transcript 1 and Transcript 2 expression was generated by CRISPR/Cas9 to determine their role in male reproduction. Results showed that 1700121C10Rik-KO male mice were fully fertile with approximately standard testis size, testicular histology, sperm production, sperm morphology, sperm motility, and induction of acrosome reaction. Thus, we conclude that both the testis-specific 1700121C10Rik-produced lncRNAs are dispensable for male fertility in mice under standard laboratory conditions.


Asunto(s)
Fertilidad/genética , Infertilidad Masculina/genética , ARN Largo no Codificante/genética , Espermatogénesis/genética , Testículo/metabolismo , Animales , Infertilidad Masculina/metabolismo , Masculino , Ratones , Ratones Noqueados , ARN Largo no Codificante/metabolismo , Motilidad Espermática/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...