Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 69
Filtrer
1.
Cell Biochem Funct ; 42(2): e3982, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38488412

RÉSUMÉ

Bone Morphogenetic Protein 2 (BMP2), a member of the Transforming Growth Factor-ß (TGF-ß) super family of proteins and is instrumental in the repair of fractures. The synthesis of BMP2 involves extensive post-translational processing and several studies have demonstrated the abysmally low production of rhBMP2 in eukaryotic systems, which may be due to the short half-life of the bioactive protein. Consequently, production costs of rhBMP2 are quite high, limiting its availability to the general populace. Therefore, there is an urgent need to identify better in-vitro systems for large scale production of rhBMP2. In the present study, we have carried out a comparative analysis of rhBMP2 production by the conventionally used Chinese Hamster ovarian cells (CHO) and goat mammary epithelial cells (GMEC), upon transfection with appropriate construct. Udder gland cells are highly secretory, and we reasoned that such cells may serve as a better in-vitro model for large scale production of rhBMP2. Our results indicated that the synthesis and secretion of bioactive rhBMP2 by goat mammary epithelial cells was significantly higher as compared to that by CHO-K1 cells. Our results provide strong evidence that GMECs may serve as a better alternative to other mammalian cells used for therapeutic protein production.


Sujet(s)
Protéine morphogénétique osseuse de type 2 , Capra , Cricetinae , Animaux , Humains , Protéine morphogénétique osseuse de type 2/pharmacologie , Cricetulus , Facteur de croissance transformant bêta , Protéines recombinantes/pharmacologie , Cellules épithéliales
2.
Biochim Biophys Acta Gene Regul Mech ; 1867(1): 194994, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37956710

RÉSUMÉ

The generation of spermatozoa from developing germ cells through mitotic and meiotic divisions is a highly regulated and complex process. Any defect in this process, may lead to subfertility/infertility. The role of different transcripts (mRNA/miRNA/lncRNA) in regulation of the pre-meiotic, meiotic, and post-meiotic stages of spermatogenesis are being proposed based on various multiomics based approaches. Such differential gene-expression is regulated by promoter elements that are activated in a stage specific manner. To determine the role of these differentially expressed transcripts in the process of meiosis, a robust post-meiotic germ cell specific promoter is required. In the present study, we have isolated and characterized the expression of the mouse Proacrosin, SP10, and ELP promoters for driving post-meiotic germ cell specific gene-expression. Promoter regions of all these 3 genes were isolated and cloned to generate mammalian expression vector. The transgene expression in post-meiotic germ cells was assessed in mice using the testicular electroporation method in vitro as well as in vivo, using above promoters. It was also validated in goat seminiferous tubules, in vitro. We have also carried out a comparative analysis of the strength of these promoters to confirm their robustness that indicated Proacrosin to be the most robust promoter that can be useful for diving post-meiotic germ cells specific gene-expression. These promoters can be used to alter gene-expression specifically in post-meiotic germ cells for deciphering the role(s) of germ cell genes in spermatogenic progression or for expressing various genome editing tools for engineering the germ cell genome to understand basis of subfertility/infertility.


Sujet(s)
Cellules germinales , Infertilité , Mâle , Souris , Animaux , Régions promotrices (génétique)/génétique , Expression des gènes , Génomique , Mammifères
3.
Reprod Sci ; 31(4): 942-965, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38036863

RÉSUMÉ

Spermatogenesis is a complex process of germ cell division and differentiation that involves extensive cross-talk between the developing germ cells and the somatic testicular cells. Defective endocrine signaling and/or intrinsic defects within the testes can adversely affect spermatogenic progression, leading to subfertility/infertility. In recent years, male infertility has been recognized as a global public health concern, and research over the last few decades has elucidated the complex etiology of male infertility. Congenital reproductive abnormalities, genetic mutations, and endocrine/metabolic dysfunction have been demonstrated to be involved in infertility/subfertility in males. Furthermore, acquired factors like exposure to environmental toxicants and lifestyle-related disorders such as illicit use of psychoactive drugs have been shown to adversely affect spermatogenesis. Despite the large body of available scientific literature on the etiology of male infertility, a substantial proportion of infertility cases are idiopathic in nature, with no known cause. The inability to treat such idiopathic cases stems from poor knowledge about the complex regulation of spermatogenesis. Emerging scientific evidence indicates that defective functioning of testicular Sertoli cells (Sc) may be an underlying cause of infertility/subfertility in males. Sc plays an indispensable role in regulating spermatogenesis, and impaired functional maturation of Sc has been shown to affect fertility in animal models as well as humans, suggesting abnormal Sc as a potential underlying cause of reproductive insufficiency/failure in such cases of unexplained infertility. This review summarizes the major causes of infertility/subfertility in males, with an emphasis on infertility due to dysregulated Sc function.


Sujet(s)
Infertilité masculine , Testicule , Animaux , Mâle , Humains , Infertilité masculine/génétique , Cellules de Sertoli , Spermatogenèse/génétique , Fécondité
4.
Animals (Basel) ; 13(5)2023 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-36899741

RÉSUMÉ

The effect of breed on milk components-fat, protein, lactose, and water-has been observed to be significant. As fat is one of the major price-determining factors for milk, exploring the variations in fat QTLs across breeds would shed light on the variable fat content in their milk. Here, on whole-genome sequencing, 25 differentially expressed hub or bottleneck fat QTLs were explored for variations across indigenous breeds. Out of these, 20 genes were identified as having nonsynonymous substitutions. A fixed SNP pattern in high-milk-yielding breeds in comparison to low-milk-yielding breeds was identified in the genes GHR, TLR4, LPIN1, CACNA1C, ZBTB16, ITGA1, ANK1, and NTG5E and, vice versa, in the genes MFGE8, FGF2, TLR4, LPIN1, NUP98, PTK2, ZTB16, DDIT3, and NT5E. The identified SNPs were ratified by pyrosequencing to prove that key differences exist in fat QTLs between the high- and low-milk-yielding breeds.

5.
Microbiol Spectr ; : e0313522, 2023 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-36853003

RÉSUMÉ

Leptospirosis, a bacterial zoonosis caused by pathogenic Leptospira spp., is prevalent worldwide and has become a serious threat in recent years. Limited understanding of Leptospira pathogenesis and host response has hampered the development of effective vaccine and diagnostics. Although Leptospira is phagocytosed by innate immune cells, it resists its destruction, and the evading mechanism involved is unclear. In the present study, we used an integrative multi-omics approach to identify the critical molecular factors of Leptospira involved in pathogenesis during interaction with human macrophages. Transcriptomic and proteomic analyses were performed at 24 h postinfection of human macrophages (phorbol-12-myristate-13-acetate differentiated THP-1 cells) with the pathogenic Leptospira interrogans serovar Icterohaemorrhagiae strain RGA (LEPIRGA). Our results identified a total of 1,528 transcripts and 871 proteins that were significantly expressed with an adjusted P value of <0.05. The correlations between the transcriptomic and proteomic data were above average (r = 0.844), suggesting the role of the posttranscriptional processes during host interaction. The conjoint analysis revealed the expression of several virulence-associated proteins such as adhesins, invasins, and secretory and chemotaxis proteins that might be involved in various processes of attachment and invasion and as effectors during pathogenesis in the host. Further, the interaction of bacteria with the host cell (macrophages) was a major factor in the differential expression of these proteins. Finally, eight common differentially expressed RNA-protein pairs, predicted as virulent, outer membrane/extracellular proteins were validated by quantitative PCR. This is the first report using integrated multi-omics approach to identify critical factors involved in Leptospira pathogenesis. Validation of these critical factors may lead to the identification of target antigens for the development of improved diagnostics and vaccines against leptospirosis. IMPORTANCE Leptospirosis is a zoonotic disease of global importance. It is caused by a Gram-negative bacterial spirochete of the genus Leptospira. The current challenge is to detect the infection at early stage for treatment or to develop potent vaccines that can induce cross-protection against various pathogenic serovars. Understanding host-pathogen interactions is important to identify the critical factors involved in pathogenesis and host defense for developing improved vaccines and diagnostics. Utilizing an integrated multi-omics approach, our study provides important insight into the interaction of Leptospira with human macrophages and identifies a few critical factors (such as virulence-associated proteins) involved in pathogenesis. These factors can be exploited for the development of novel tools for the detection, treatment, or prevention of leptospirosis.

6.
J Mol Endocrinol ; 69(3): 431-444, 2022 10 01.
Article de Anglais | MEDLINE | ID: mdl-35917434

RÉSUMÉ

Endometrial cancer is the fourth most common malignancy in women and the precursor lesion is endometrial hyperplasia. HOXA10 is a transcription factor that plays key roles in endometrial functions such as the endowment of receptivity, embryo implantation, and trophoblast invasion. Herein, using testicular transgenesis, we developed transgenic mice that expressed a shRNA against HOXA10 and there was a nearly 70% reduction in the expression of HOXA10 in these animals. We observed that downregulation of HOXA10 led to the development of endometrial hyperplasia in the young animals (3 months), and as they aged (>1 year), most animals developed well-differentiated endometrial adenocarcinoma. In the endometrium of animals with reduced HOXA10, there was increased proliferation and elevated levels of ERα and ERß. In parallel, there was increased expression of Wnt4 and ß-Catenin, SOX9, and YAP1. We propose that chronic reduction in HOXA10 expression disrupts multiple pathways in the uterus that aids in the development of endometrial hyperplasia which progresses to endometrial cancer with age.


Sujet(s)
Hyperplasie endométriale , Tumeurs de l'endomètre , Animaux , Implantation embryonnaire/physiologie , Hyperplasie endométriale/génétique , Hyperplasie endométriale/métabolisme , Tumeurs de l'endomètre/anatomopathologie , Endomètre/métabolisme , Femelle , Protéines à homéodomaines A10 , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Humains , Souris
7.
Biotechnol Bioeng ; 119(11): 3275-3283, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35896515

RÉSUMÉ

Transfection of nucleic acid molecules into mammalian cells can be facilitated using viral vectors, electroporation, or biocompatible cationic materials. However, safety issues and the requirement of specialized equipment limits the use of viral vectors and physical methods of transfection like electroporation and microinjection, respectively. Biocompatible cationic lipids and polymers like branched-polyethyleneimine (bPEI) have a wide transfection range and are user-friendly in most applications. However, bPEI exhibits low transfection efficiency in most cell types. In the present work, we have crosslinked the hexanoyl group to bPEI using anhydride chemistry to enhance its efficiency as a transfection reagent. The efficient association of hexanoyl group to bPEI was assessed using Fourier-transform infrared spectroscopy and other physicochemical methods. Hexanoyl-modified bPEI (FA6-bPEI) was found to exhibit significantly enhanced transfection efficiency in both cell lines and cultured primary cells, as compared to native bPEI and the commercially available transfection reagent, Lipofectamine 3000. Furthermore, our in vitro studies indicated that FA6-bPEI can be used for robust transfection for increased production of therapeutic proteins in a cell culture-based system. These results suggested that hexanoyl-modified bPEI can serve as an efficient transfection reagent for studies on hard-to-transfect cells and for enhanced production of therapeutic proteins in vitro.


Sujet(s)
Acides nucléiques , Polyéthylèneimine , Anhydrides , Animaux , Matériaux biocompatibles , Lignée cellulaire , ADN/métabolisme , Mammifères/métabolisme , Acides nucléiques/métabolisme , Polyéthylèneimine/composition chimique , Polyéthylèneimine/métabolisme , Polymères/composition chimique , Transfection
8.
Cell Mol Life Sci ; 79(3): 136, 2022 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-35181820

RÉSUMÉ

Sertoli cells (Sc) are the sole target of follicle-stimulating hormone (FSH) in the testis and attain functional maturation post-birth to significantly augment germ cell (Gc) division and differentiation at puberty. Despite having an operational microRNA (miRNA) machinery, limited information is available on miRNA-mediated regulation of Sc maturation and male fertility. We have shown before that miR-92a-3p levels decline in pubertal rat Sc. In response to FSH treatment, the expressions of FSH Receptor, Claudin11 and Klf4 were found to be elevated in pubertal rat Sc coinciding with our finding of FSH-induced decline in miR-92a-3p levels. To investigate the association of miR-92a-3p and spermatogenesis, we generated transgenic mice where such pubertal decline of miR-92a-3p was prevented by its overexpression in pubertal Sc under proximal Rhox5 promoter, which is known to be activated specifically at puberty, in Sc. Our in vivo observations provided substantial evidence that FSH-induced decline in miR-92a-3p expression during Sc maturation acts as an essential prerequisite for the pubertal onset of spermatogenesis. Elevated expression of miR-92a-3p in post-pubertal testes results into functionally compromised Sc, leading to impairment of the blood-testis barrier formation and apoptosis of pre-meiotic Gc, ultimately culminating into infertility. Collectively, our data suggest that regulation of miR-92a-3p expression is crucial for Sc-mediated induction of active spermatogenesis at puberty and regulation of male fertility.


Sujet(s)
Différenciation cellulaire , Fécondité , Hormone folliculostimulante/pharmacologie , Cellules germinales/cytologie , microARN/génétique , Cellules de Sertoli/cytologie , Testicule/cytologie , Animaux , Femelle , Cellules germinales/effets des médicaments et des substances chimiques , Cellules germinales/métabolisme , Hormones/pharmacologie , Mâle , Souris , Souris transgéniques , Rats , Rat Wistar , Récepteur FSH/génétique , Récepteur FSH/métabolisme , Cellules de Sertoli/effets des médicaments et des substances chimiques , Cellules de Sertoli/métabolisme , Maturation sexuelle , Spermatogenèse , Testicule/effets des médicaments et des substances chimiques , Testicule/métabolisme
9.
Mol Ther Nucleic Acids ; 26: 192-207, 2021 Dec 03.
Article de Anglais | MEDLINE | ID: mdl-34513304

RÉSUMÉ

A major change in the transcriptome of testicular Sertoli cells (Scs) at the onset of puberty enables them to induce robust spermatogenesis. Through comprehensive literature mining, we generated a list of genes crucial for Sc functioning and computationally predicted the microRNAs regulating them. Differential expression analysis of microRNAs in infant and pubertal rat Scs showed that miR-382-3p levels decline significantly in pubertal Scs. Interestingly, miR-382-3p was found to regulate genes like Ar and Wt1, which are crucial for functional competence of Scs. We generated a transgenic (Tg) mouse model in which pubertal decline of miR-382-3p was prevented by its overexpression in pubertal Scs. Elevated miR-382-3p restricted the functional maturation of Scs at puberty, leading to infertility. Prevention of decline in miR-382-3p expression in pubertal Scs was responsible for defective blood-testis barrier (BTB) formation, severe testicular defects, low epididymal sperm counts and loss of fertility in these mice. This provided substantial evidence that decline in levels of miR-382-3p at puberty is the essential trigger for onset of robust spermatogenesis at puberty. Hence, sustained high levels of miR-382-3p in pubertal Scs could be one of the underlying causes of idiopathic male infertility and should be considered for diagnosis and treatment of infertility.

10.
Bioengineered ; 12(1): 4407-4419, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34436976

RÉSUMÉ

Widespread infection due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has led to a global pandemic. Currently, various approaches are being taken up to develop vaccines and therapeutics to treat SARS-CoV2 infection. Consequently, the S protein has become an important target protein for developing vaccines and therapeutics against SARS-CoV2. However, the highly infective nature of SARS-CoV2 restricts experimentation with the virus to highly secure BSL3 facilities. The availability of fusion-enabled, nonreplicating, and nonbiohazardous mimics of SARS-CoV2 virus fusion, containing the viral S or S and M protein in their native conformation on mammalian cells, can serve as a useful substitute for studying viral fusion for testing various inhibitors of viral fusion. This would avoid the use of the BSL3 facility for fusion studies required to develop therapeutics. In the present study, we have developed SARS-CoV2 virus fusion mimics (SCFMs) using mammalian cells transfected with constructs coding for S or S and M protein. The fusogenic property of the mimic(s) and their interaction with the functional human ACE2 receptors was confirmed experimentally. We have also shown that such mimics can easily be used in an inhibition assay. These mimic(s) can be easily prepared on a large scale, and such SCFMs can serve as an invaluable resource for viral fusion inhibition assays and in vitro screening of antiviral agents, which can be shared/handled between labs/facilities without worrying about any biohazard while working under routine laboratory conditions, avoiding the use of BSL3 laboratory.Abbreviations :SCFM: SARS-CoV2 Virus Fusion Mimic; ACE2: Angiotensin-Converting Enzyme 2; hACE2: Human Angiotensin-Converting enzyme 2; MEF: Mouse Embryonic Fibroblasts; HBSS: Hanks Balanced Salt Solution; FBS: Fetal Bovine Serum.


Sujet(s)
Anticorps neutralisants/pharmacologie , Confinement de risques biologiques/méthodes , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/antagonistes et inhibiteurs , Protéines de la matrice virale/antagonistes et inhibiteurs , Pénétration virale/effets des médicaments et des substances chimiques , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , Animaux , Chlorocebus aethiops , Embryon de mammifère , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/virologie , Expression des gènes , Gènes rapporteurs , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Cellules HEK293 , Humains , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Cellules MCF-7 , Souris , Mimétisme moléculaire , Plasmides/composition chimique , Plasmides/métabolisme , Culture de cellules primaires , Liaison aux protéines , Récepteurs viraux/génétique , Récepteurs viraux/métabolisme , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/métabolisme , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme , Transfection , Cellules Vero , Protéines de la matrice virale/génétique , Protéines de la matrice virale/métabolisme
11.
Biochem Biophys Res Commun ; 569: 100-105, 2021 09 10.
Article de Anglais | MEDLINE | ID: mdl-34237428

RÉSUMÉ

Follicle Stimulating Hormone (FSH) acts via FSH-Receptor (FSH-R) by employing cAMP as the dominant secondary messenger in testicular Sertoli cells (Sc) to support spermatogenesis. Binding of FSH to FSH-R, results the recruitment of the intracellular GTP binding proteins, either stimulatory Gαs or inhibitory Gαi that in turn regulate cAMP production in Sc. The cytosolic concentration of cAMP being generated by FSH-R thereafter critically determines the downstream fate of the FSH signalling. The pleiotropic action of FSH due to differential cAMP output during functional maturation of Sc has been well studied. However, the developmental and cellular regulation of the Gα proteins associated with FSH-R is poorly understood in Sc. In the present study, we report the differential transcriptional modulation of the Gα subunit genes by FSH mediated cAMP signalling in neonatal and pubertal rat Sc. Our data suggested that unlike in neonatal Sc, both the basal and FSH/forskolin induced expression of Gαs, Gαi-1, Gαi-2 and Gαi-3 transcripts was significantly (p < 0.05) up-regulated in pubertal Sc. Further investigations involving treatment of Sc with selective Gαi inhibitor pertussis toxin, confirmed the elevated expression of Gi subunits in pubertal Sc. Collectively our results indicated that the high level of Gαi subunits serves as a negative regulator to optimize cAMP production in pubertal Sc.


Sujet(s)
AMP cyclique/métabolisme , Hormone folliculostimulante/pharmacologie , Sous-unités alpha Gi-Go des protéines G/métabolisme , Cellules de Sertoli/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Animaux nouveau-nés , Cellules cultivées , Colforsine/pharmacologie , Hormone folliculostimulante/métabolisme , Sous-unités alpha Gi-Go des protéines G/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Mâle , Toxine pertussique/pharmacologie , Liaison aux protéines , Rat Wistar , Récepteur FSH/génétique , Récepteur FSH/métabolisme , RT-PCR , Cellules de Sertoli/cytologie , Cellules de Sertoli/métabolisme , Maturation sexuelle/physiologie , Spermatogenèse/effets des médicaments et des substances chimiques , Spermatogenèse/génétique
12.
Cell Biol Int ; 45(8): 1720-1732, 2021 Aug.
Article de Anglais | MEDLINE | ID: mdl-33847415

RÉSUMÉ

Keratin 8/18, the predominant keratin pair of simple epithelia, is known to be aberrantly expressed in several squamous cell carcinomas (SCCs), where its expression is often correlated with increased invasion, neoplastic progression, and poor prognosis. The majority of keratin 8/18 structural and regulatory functions are governed by posttranslational modifications, particularly phosphorylation. Apart from filament reorganization, cellular processes including cell cycle, cell growth, cellular stress, and apoptosis are known to be orchestrated by K8 phosphorylation at specific residues in the head and tail domains. Even though deregulation of K8 phosphorylation at two significant sites (Serine73 /Serine431 ) has been implicated in neoplastic progression of SCCs by various in vitro studies, including ours, it is reported to be highly context-dependent. Therefore, to delineate the precise role of Kereatin 8 phosphorylation in cancer initiation and progression, we have developed the tissue-specific transgenic mouse model expressing Keratin 8 wild type and phosphodead mutants under Keratin 14 promoter. Subjecting these mice to 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate-mediated skin carcinogenesis revealed that Keratin 8 phosphorylation may lead to an early onset of tumors compared to Keratin 8 wild-type expressing mice. Conclusively, the transgenic mouse model developed in the present study ascertained a positive impact of Keratin 8 phosphorylation on the neoplastic transformation of skin-squamous cells.


Sujet(s)
Carcinogenèse/métabolisme , Kératine-8/métabolisme , Mutation/physiologie , Tumeurs cutanées/métabolisme , Animaux , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Électroporation/méthodes , Cellules HEK293 , Humains , Kératine-8/génétique , Mâle , Souris , Souris transgéniques , Phosphorylation/physiologie , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie
13.
Biochim Biophys Acta Mol Basis Dis ; 1867(4): 166050, 2021 04 01.
Article de Anglais | MEDLINE | ID: mdl-33359696

RÉSUMÉ

Obesity induced insulin resistance is primarily regulated by the inhibitory phosphorylation of peroxisome proliferator-activated receptor γ at serine 273 (PPARγS273) which has been shown to be regulated by MEK and ERK. An upstream regulatory molecule of this pathway could be a therapeutic option. Here we analyzed the involvement of Fetuin-A (FetA), a key hepato-adipokine implicated in insulin resistance, as an upstream regulator molecule for the regulation of PPARγ inhibitory phosphorylation. Mice fed with standard diet (SD), high fat diet (HFD) and HFD with FetA knockdown (HFD-FetAKD) were used to examine the role of FetA on PPARγS273 phosphorylation in adipocytes. The mechanism of regulation and its effect on skeletal muscle were studied using primary adipocytes, 3T3-L1 (preadipocyte) and C2C12 (myotube) cell lines. Increased FetA in HFD mice strongly correlated with augmentation of PPARγS273 phosphorylation in inflamed adipocytes while knockdown of FetA suppressed it. This effect of FetA was mediated through the activation of Ras which in turn activated MEK and ERK. On addressing how FetA could stimulate activation of Ras, we found that FetA triggered TNFα in inflamed adipocytes which induced Ras activation. The ensuing sharp fall in adiponectin level attenuated AMPK activation in skeletal muscle cells affecting mitochondrial ATP production. Our data reveal the essential role of FetA induced activation of Ras in regulating PPARγ inhibitory phosphorylation through Ras-MEK-ERK pathway which downregulates adiponectin disrupting skeletal muscle mitochondrial bioenergetics. Thus, FetA mediated PPARγ inactivation has adverse consequences upon adipocyte-myocyte crosstalk leading to disruption of energy homeostasis and loss of insulin sensitivity.


Sujet(s)
Insulinorésistance , Système de signalisation des MAP kinases , Obésité/métabolisme , Récepteur PPAR gamma/métabolisme , alpha-2-HS-glycoprotéine/métabolisme , Cellules 3T3-L1 , Animaux , Cellules cultivées , Métabolisme énergétique , Mâle , Souris , Mitochondries/métabolisme , Phosphorylation
14.
Andrology ; 9(2): 689-699, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33145986

RÉSUMÉ

BACKGROUND: Infertility has become a global phenomenon and constantly declining sperm count in males in modern world pose a major threat to procreation of humans. Male fertility is critically dependent on proper functioning of testicular Sertoli cells. Defective Sertoli cell proliferation and/or impaired functional maturation may be one of the underlying causes of idiopathic male infertility. Using high-throughput "omics" approach, we found binding sites for homeobox transcription factor MEIS1 on the promoters of several genes up-regulated in pubertal (mature) Sertoli cells, indicating that MEIS1 may be crucial for Sertoli cell-mediated regulation of spermatogenesis at and after puberty. OBJECTIVE: To decipher the role of transcription factor MEIS1 in Sertoli cell maturation and spermatogenesis. MATERIALS AND METHODS: Sc-specific Meis1 knockdown (KD) transgenic mice were generated using pronuclear microinjection. Morphometric and histological analysis of the testes from transgenic mice was performed to identify defects in spermatogenesis. Epididymal sperm count and litter size were analyzed to determine the effect of Meis1 knockdown on fertility. RESULTS: Sertoli cell (Sc)-specific Meis1 KD led to massive germ cell loss due to apoptosis and impaired spermatogenesis. Unlike normal pubertal Sc, the levels of SOX9 in pubertal Sc of Meis1 KD were significantly high, like immature Sc. A significant reduction in epididymal sperm count was observed in these mice. The mice were found to be infertile or sub-fertile (with reduced litter size), depending on the extent of Meis1 inhibition. DISCUSSION: The results of this study demonstrated for the first time, a role of Meis1 in Sc maturation and normal spermatogenic progression. Inhibition of Meis1 in Sc was associated with deregulated spermatogenesis and a consequent decline in fertility of the transgenic mice. CONCLUSIONS: Our results provided substantial evidence that suboptimal Meis1 expression in Sc may be one of the underlying causes of idiopathic infertility.


Sujet(s)
Fécondité/physiologie , Protéine du site-1 d'intégration des virus myéloïdes écotropiques/physiologie , Cellules de Sertoli/physiologie , Animaux , Fécondité/génétique , Techniques de knock-down de gènes , Mâle , Souris transgéniques , Protéine du site-1 d'intégration des virus myéloïdes écotropiques/génétique , Cellules de Sertoli/métabolisme , Spermatogenèse/génétique , Spermatogenèse/physiologie
15.
Mol Hum Reprod ; 26(10): 760-772, 2020 10 01.
Article de Anglais | MEDLINE | ID: mdl-32687199

RÉSUMÉ

The alarming decline in sperm count has become a global concern in the recent decades. The division and differentiation of male germ cells (Gc) into sperm are governed by Sertoli cells (Sc) upon their functional maturation during puberty. However, the roles of genes regulating pubertal maturation of Sc have not been fully determined. We have observed that Tetraspanin 8 (Tspan8) is down-regulated in Sc during puberty in rats. However, there has been no in vivo evidence for a causal link between the down-regulation of Tspan8 expression and the onset of spermatogenesis as yet. To investigate this, we generated a novel transgenic (Tg) rat, in which the natural down-regulation of Tspan8 was prevented specifically in Sc from puberty up to adulthood. Adult Tg male rats showed around 98% reduction in sperm count despite having a similar level of serum testosterone (T) as the controls. Functional maturation of Sc was impaired as indicated by elevated levels of Amh and low levels of Kitlg and Claudin11 transcripts. The integrity of the blood testis barrier was compromised due to poor expression of Gja1 and Gc apoptosis was discernible. This effect was due to a significant rise in both Mmp7 and phospho P38 MAPK in Tg rat testis. Taken together, we demonstrated that the natural down-regulation of Tspan8 in Sc during puberty is a prerequisite for establishing male fertility. This study divulges one of the aetiologies of certain forms of idiopathic male infertility where somatic cell defect, but not hormonal deficiency, is responsible for impaired spermatogenesis.


Sujet(s)
Infertilité masculine/génétique , Cellules de Sertoli/métabolisme , Maturation sexuelle/génétique , Tétraspanines/génétique , Animaux , Régulation négative/génétique , Femelle , Fécondité/génétique , Régulation de l'expression des gènes au cours du développement , Infertilité masculine/métabolisme , Mâle , Grossesse , Rats , Rats transgéniques , Rat Wistar , Testicule/métabolisme , Tétraspanines/métabolisme
16.
Mol Reprod Dev ; 86(11): 1505-1530, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31368614

RÉSUMÉ

The term "Puberty", socially known as "Adolescence" is the transitional period from juvenile life to adulthood with functional maturation of gonads and genital organs. In this process, some remarkable developmental changes occur in morphology, physiology, and behavior leading to reproductive competence. Despite sufficient levels of gonadotropins (luteinizing hormone [LH] and follicle-stimulating hormone [FSH]), robust spermatogenesis is not initiated during infancy in primates due to the immaturity of testicular Sertoli cells. Recent studies suggest that developmental competence augmenting functional activities of receptors for androgen and FSH is acquired by Sertoli cells somewhere during the prolonged hypo-gonadotropic juvenile period. This juvenile phase is terminated with the re-awakening of hypothalamic Kisspeptin/Neurokinin B/Dynorphin neurons which induce the release of the gonadotropin-releasing hormone leading to reactivation of the hypothalamo-pituitary-testicular axis at puberty. During this period of pubertal development, FSH and LH facilitate further maturation of testicular cells (Sertoli cells and Leydig cells) triggering robust differentiation of the spermatogonial cells, ensuing the spermatogenic onset. This review aims to precisely address the evolving concepts of the pubertal regulation of hormone production with the corresponding cooperation of testicular cells for the initiation of robust spermatogenesis, which can be truly called "testicular puberty."


Sujet(s)
Hormone folliculostimulante/métabolisme , Hormone de libération des gonadotrophines/métabolisme , Cellules de Leydig/métabolisme , Hormone lutéinisante/métabolisme , Cellules de Sertoli/métabolisme , Maturation sexuelle/physiologie , Spermatogenèse/physiologie , Animaux , Cellules de Leydig/cytologie , Mâle , Primates , Cellules de Sertoli/cytologie
17.
Sci Rep ; 9(1): 11458, 2019 08 07.
Article de Anglais | MEDLINE | ID: mdl-31391487

RÉSUMÉ

An alarming decline in sperm count of men from several countries has become a major concern for the world community. Hormones act on testicular Sertoli cells (Sc) to regulate male fertility by governing the division and differentiation of germ cells (Gc). However, there is a limited knowledge about Sc specific gene(s) regulating the spermatogenic output of the testis. Sclerostin domain-containing 1 protein (Sostdc1) is a dual BMP/Wnt regulator is predominantly expressed in the Sc of infant testes which hardly show any sign of spermatogenesis. In order to investigate the role of Sostdc1 in spermatogenic regulation, we have generated transgenic (Tg) rats which induced persistent expression of Sostdc1 in mature Sc causing reduced sperm counts. Although Sc specific Sostdc1 did not affect the function of either Sc or Leydig cells (Lc) in the adult testis of Tg rat, we observed a selective augmentation of the BMP target genes via activated phospho smad 1/5/8 signaling in Gc leading to apoptosis. Here, for the first time, we have demonstrated that Sostdc1 is a negative regulator of spermatogenesis, and provided substantial evidence that down regulation of Sostdc1 during puberty is critically essential for quantitatively and qualitatively normal spermatogenesis governing male fertility.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Oligospermie/anatomopathologie , Cellules de Sertoli/métabolisme , Spermatogenèse/physiologie , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Apoptose/physiologie , Biopsie , Protéines morphogénétiques osseuses/métabolisme , Études cas-témoins , Jeux de données comme sujet , Régulation négative/physiologie , Cellules de Leydig/métabolisme , Mâle , Modèles animaux , Oligospermie/génétique , Rats , Rats transgéniques , Maturation sexuelle/physiologie , Transduction du signal/physiologie , Testicule/cytologie , Testicule/anatomopathologie , Analyse sur puce à tissus
18.
Mol Cell Endocrinol ; 482: 70-80, 2019 02 15.
Article de Anglais | MEDLINE | ID: mdl-30579957

RÉSUMÉ

The synergistic actions of Testosterone (T) and FSH via testicular Sertoli cells (Sc) regulate male fertility. We have previously reported that the actions of these hormones (T and FSH) in infant monkey testes are restricted only to the expansion of Sc and spermatogonial cells. The robust differentiation of male Germ cells (Gc) occurs after pubertal maturation of testis. The present study was aimed to investigate the molecular basis of the synergy between T and FSH action in pubertal primate (Macaca mulatta) Sc. Using primary Sc culture, we here have demonstrated that T (but not FSH) downregulated AMH and Inhibin-ß-B (INHBB) mRNAs in pubertal Sc. We also found that, prolonged stimulation of T in pubertal Sc significantly elevated the expression of genes involved in FSH signaling pathway like FSH-Receptor (FSHR), GNAS and RIC8B, and this was associated with a rise in cAMP production. T also augmented FSH induced expression of genes like SCF, GDNF, ABP and Transferrin (TF) in pubertal Sc. We therefore conclude that T acts in synergy with FSH signaling in pubertal Sc. Such a coordinated network of hormonal signaling in Sc may facilitate the timely onset of the first spermatogenic wave in pubertal primates and is responsible for quantitatively and qualitatively normal spermatogenesis.


Sujet(s)
Hormone folliculostimulante/pharmacologie , Récepteur FSH/génétique , Récepteur FSH/métabolisme , Cellules de Sertoli/cytologie , Testostérone/pharmacologie , Animaux , Cellules cultivées , Sous-unités bêta de l'inhibine/génétique , Macaca mulatta , Mâle , Cellules de Sertoli/effets des médicaments et des substances chimiques , Cellules de Sertoli/métabolisme , Maturation sexuelle , Transduction du signal/effets des médicaments et des substances chimiques , Régulation positive
19.
Appl Microbiol Biotechnol ; 102(14): 6221-6234, 2018 Jul.
Article de Anglais | MEDLINE | ID: mdl-29855689

RÉSUMÉ

Protein expression in the milk of transgenic farmed animals offers a cost-effective system for producing therapeutics. However, transgenesis in farmed animals is not only cumbersome but also involves risk of potential hazard by germline gene integration, due to interruptions caused by the transgene in the native genome. Avoiding germline gene integration, we have delivered buffalo ß-casein promoter-driven transgene construct entrapped in virosomes directly in the milk gland through intraductal perfusion delivery. Virosomes were generated from purified Sendai viral membrane, containing hemagglutinin-neuraminidase (HN) and fusion factor (F) proteins on surface (HNF-Virosomes) which initiate membrane fusion, devoid of any viral nucleic acids. Intraductal delivery of HNF-Virosomes predominantly transfected luminal epithelial cells lining the milk duct and buffalo ß-casein promoter of the construct ensured mammary luminal epithelial cell specific expression of the transgene. Mammary epithelial cells expressed EGFP at lactation when egfp was used as a transgene. Similarly, human interferon-γ (hIFN-γ) was expressed in the mammary gland as well as in the milk when hIFN-γ was used as a transgene. This combinatorial approach of using Sendai viral membrane-derived virosomes for entrapment and delivery of the transgene and using buffalo ß-casein promoter for mammary gland specific gene expression provided a better option for generating therapeutic proteins in milk, bypassing germline gene integration avoiding risks associated with animal bioreactor generated through germline gene integration.


Sujet(s)
Biothérapie/méthodes , Buffles/génétique , Expression des gènes/génétique , Lactation/génétique , Glandes mammaires animales/métabolisme , Lait/composition chimique , Transgènes/génétique , Animaux , Caséines/génétique , Femelle , Humains , Régions promotrices (génétique)/génétique , Virus Sendai/génétique
20.
Biochem Biophys Res Commun ; 501(3): 771-778, 2018 06 27.
Article de Anglais | MEDLINE | ID: mdl-29763604

RÉSUMÉ

Accumulation and polarization of anti-inflammatory M2 to proinflammatory M1 macrophage in the adipose tissue of obese diabetic mice is an important pathogenic signature. It worsens lipid induced inflammation and insulin resistance. Here we demonstrate that a small molecule, a peroxyvanadate compound i.e. DmpzH [VO(O2)2 (dmpz)] or dmp, could robustly decrease macrophage infiltration, accumulation and their polarization in high fat diet (HFD) induced obese diabetic mice. In searching the underlying mechanism it was revealed that SIRT1 level was strikingly low in the inflamed adipose tissue of HFD mice as compared to mice fed with standard diet (SD). Administration of dmp markedly increased SIRT1 level by inducing its gene expression with a consequent decrease in macrophage population. Elevation of SIRT1 coincided with the decrease of MCP1, Fetuin-A (FetA) and IFNγ. Since MCP1 and FetA drive macrophage to inflamed adipose tissue and IFNγ promotes M2 to M1 transformation, both recruitment and M1 induced inflammation were found to be significantly repressed by dmp. In addressing the question about how dmp induced excess SIRT1 could reduce MCP1, FetA and IFNγ levels, we found that it was due to the inactivation of NFκB because of its deacetylation by SIRT1. Since NFκB is the transcriptional regulator of these molecules, their expressions were significantly suppressed and that caused sharp decline in macrophage recruitment and their polarity to M1. This effected a marked fall in proinflammatory cytokine level which significantly improved insulin sensitivity. dmp is likely to be the first molecule that rescues inflammatory burden contributed by macrophage in obese diabetic mice adipose tissue which causes significant increase in insulin sensitivity therefore it may be a meaningful choice to treat type 2 diabetes.


Sujet(s)
Diabète expérimental/complications , Diabète expérimental/traitement médicamenteux , Insulinorésistance , Macrophages/effets des médicaments et des substances chimiques , Obésité/complications , Obésité/traitement médicamenteux , Vanadates/usage thérapeutique , Animaux , Polarité de la cellule/effets des médicaments et des substances chimiques , Diabète expérimental/anatomopathologie , Diabète de type 2/complications , Diabète de type 2/traitement médicamenteux , Diabète de type 2/anatomopathologie , Inflammation/complications , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Macrophages/anatomopathologie , Mâle , Souris , Souris de lignée BALB C , Souris obèse , Obésité/anatomopathologie , Cellules RAW 264.7
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...