Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1248907, 2024.
Article in English | MEDLINE | ID: mdl-38720893

ABSTRACT

Introduction: Sepsis remains a major cause of death in Intensive Care Units. Sepsis is a life-threatening multi-organ dysfunction caused by a dysregulated systemic inflammatory response. Pattern recognition receptors, such as TLRs and NLRs contribute to innate immune responses. Upon activation, some NLRs form multimeric protein complexes in the cytoplasm termed "inflammasomes" which induce gasdermin d-mediated pyroptotic cell death and the release of mature forms of IL-1ß and IL-18. The NLRP6 inflammasome is documented to be both a positive and a negative regulator of host defense in distinct infectious diseases. However, the role of NLRP6 in polymicrobial sepsis remains elusive. Methods: We have used NLRP6 KO mice and human septic spleen samples to examine the role of NLRP6 in host defense in sepsis. Results: NLRP6 KO mice display enhanced survival, reduced bacterial burden in the organs, and reduced cytokine/chemokine production. Co-housed WT and KO mice following sepsis show decreased bacterial burden in the KO mice as observed in singly housed groups. NLRP6 is upregulated in CD3, CD4, and CD8 cells of septic patients and septic mice. The KO mice showed a higher number of CD3, CD4, and CD8 positive T cell subsets and reduced T cell death in the spleen following sepsis. Furthermore, administration of recombinant IL-18, but not IL-1ß, elicited excessive inflammation and reversed the survival advantages observed in NLRP6 KO mice. Conclusion: These results unveil NLRP6 as a negative regulator of host defense during sepsis and offer novel insights for the development of new treatment strategies for sepsis.


Subject(s)
Immunity, Innate , Intracellular Signaling Peptides and Proteins , Sepsis , Animals , Female , Humans , Male , Mice , Cytokines/metabolism , Disease Models, Animal , Immunity, Innate/genetics , Inflammasomes/metabolism , Inflammasomes/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface , Sepsis/immunology , Sepsis/microbiology , Spleen/immunology
2.
Int J Mol Sci ; 25(4)2024 Feb 10.
Article in English | MEDLINE | ID: mdl-38396827

ABSTRACT

Kidney transplantation is the preferred treatment for end-stage kidney disease (ESKD). However, there is a shortage of transplantable kidneys, and donor organs can be damaged by necessary cold storage (CS). Although CS improves the viability of kidneys from deceased donors, prolonged CS negatively affects transplantation outcomes. Previously, we reported that renal proteasome function decreased after rat kidneys underwent CS followed by transplantation (CS + Tx). Here, we investigated the mechanism underlying proteasome dysfunction and the role of the proteasome in kidney graft outcome using a rat model of CS + Tx. We found that the key proteasome subunits ß5, α3, and Rpt6 are modified, and proteasome assembly is impaired. Specifically, we detected the modification and aggregation of Rpt6 after CS + Tx, and Rpt6 modification was reversed when renal extracts were treated with protein phosphatases. CS + Tx kidneys also displayed increased levels of nitrotyrosine, an indicator of peroxynitrite (a reactive oxygen species, ROS), compared to sham. Because the Rpt6 subunit appeared to aggregate, we investigated the effect of CS + Tx-mediated ROS (peroxynitrite) generation on renal proteasome assembly and function. We treated NRK cells with exogenous peroxynitrite and evaluated PAC1 (proteasome assembly chaperone), Rpt6, and ß5. Peroxynitrite induced a dose-dependent decrease in PAC1 and ß5, but Rpt6 was not affected (protein level or modification). Finally, serum creatinine increased when we inhibited the proteasome in transplanted donor rat kidneys (without CS), recapitulating the effects of CS + Tx. These findings underscore the effects of CS + Tx on renal proteasome subunit dysregulation and also highlight the significance of proteasome activity in maintaining graft function following CS + Tx.


Subject(s)
Kidney Transplantation , Rats , Animals , Kidney Transplantation/adverse effects , Proteasome Endopeptidase Complex/metabolism , Reactive Oxygen Species/metabolism , Peroxynitrous Acid/metabolism , Kidney/metabolism , Organ Preservation
4.
Mucosal Immunol ; 14(3): 574-584, 2021 05.
Article in English | MEDLINE | ID: mdl-33230225

ABSTRACT

Bacterial pneumonia is a significant cause of morbidity, mortality, and health care expenditures. Optimum neutrophil recruitment and their function are critical defense mechanisms against respiratory pathogens. The nucleotide-binding oligomerization domain-like receptor (NLRP) 6 controls gut microbiota and immune response to systemic and enteric infections. However, the importance of NLRP6 in neutrophil homeostasis following lung infection remains elusive. To investigate the role of NLRs in neutrophil homeostasis, we used Nlrp6 gene-deficient (Nlrp6-/-) mice in a model of Klebsiella pneumoniae-induced pneumonia-derived sepsis. We demonstrated that NLRP6 is critical for host survival, bacterial clearance, neutrophil influx, and CXC-chemokine production. Kp-infected Nlrp6-/- mice have reduced numbers of hematopoietic stem cells and granulocyte-monocyte progenitors but increased retention of matured neutrophils in bone marrow. Neutrophil extracellular trap (NET) formation and NET-mediated bacterial killing were also impaired in Nlrp6-/- neutrophils in vitro. Furthermore, recombinant CXCL1 rescued the impaired host defense, granulopoietic response, and NETosis in Kp-infected Nlrp6-/- mice. Using A/J background mice and co-housing experiments, our findings revealed that the susceptible phenotype of Nlrp6-/- mice is not strain-specific and gut microbiota-dependent. Taken together, these data unveil NLRP6 as a central regulator of neutrophil recruitment, generation, and function during bacterial pneumonia followed by sepsis.


Subject(s)
Klebsiella Infections/metabolism , Klebsiella pneumoniae/physiology , Macrophages/immunology , Neutrophils/immunology , Pneumonia, Bacterial/metabolism , Receptors, Cell Surface/metabolism , Sepsis/metabolism , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Extracellular Traps/metabolism , Homeostasis , Humans , Klebsiella Infections/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Activation , Pneumonia, Bacterial/immunology , Receptors, Cell Surface/genetics , Sepsis/immunology , THP-1 Cells
5.
J Dairy Res ; 87(2): 170-174, 2020 May.
Article in English | MEDLINE | ID: mdl-32482199

ABSTRACT

In this research communication we describe the DGAT1 sequence and promoter region in dairy cows and buffalo and compare the activities of DGAT1 between the two species in order to increase knowledge of the cause of milk fat variation. pGL-3 basic vectors were used to construct the reporter gene. Based on the predicted promoter region, 4 truncated plasmid vectors were constructed in cow-DGAT1 and 3 plasmid vectors in buffalo-DGAT1. Each reporter plasmid was transfected into the bovine mammary epithelial cell (BMEC), 293T cell, and CHO cells to analyze the activity using Dual-Luciferase Reporter Assay System. The results show that the region between -93 to -556 bp was essential for cow promoter activity while -84 to -590 bp was essential for buffalo promoter activity revealing these regions contain core promoter. The buffalo has higher promoter activity than cow yet it was not statistically significant. Comparison of candidate mutation K232A between cow and buffalo population revealed the presence of both the allelic population in dairy cows (lysine and alanine) however, only K (lysine) allelic amino acid was found in buffalo population. The absence of the alanine allelic population from buffalo explains the higher fat content of buffalo milk.


Subject(s)
Buffaloes/genetics , Cattle/genetics , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Lipids/biosynthesis , Milk/metabolism , Animals , CHO Cells , Cricetulus , Epithelial Cells/enzymology , Female , HEK293 Cells , Humans , Lipids/analysis , Mammary Glands, Animal/enzymology , Milk/chemistry , Promoter Regions, Genetic/genetics , Species Specificity , Transfection
6.
Biomed Res Int ; 2018: 1463732, 2018.
Article in English | MEDLINE | ID: mdl-29862252

ABSTRACT

AKT3 gene is a constituent of the serine/threonine protein kinase family and plays a crucial role in synthesis of milk fats and cholesterol by regulating activity of the sterol regulatory element binding protein (SREBP). AKT3 is highly conserved in mammals and its expression levels during the lactation periods of cattle are markedly increased. AKT3 is highly expressed in the intestine followed by mammary gland and it is also expressed in immune cells. It is involved in the TLR pathways as effectively as proinflammatory cytokines. The aims of this study were to investigate the sequences differences between buffalo and cow. Our results showed that there were substantial differences between buffalo and cow in some exons and noteworthy differences of the gene size in different regions. We also identified the important consensus sequence motifs, variation in 2000 upstream of ATG, substantial difference in the "3'UTR" region, and miRNA association in the buffalo sequences compared with the cow. In addition, genetic analyses, such as gene structure, phylogenetic tree, position of different motifs, and functional domains, were performed to establish their correlation with other species. This may indicate that a buffalo breed has potential resistance to disease, environment changes, and airborne microorganisms and some good production and reproductive traits.


Subject(s)
3' Untranslated Regions , Buffaloes , Cattle , Gene Expression Regulation, Enzymologic , Mammary Glands, Animal/enzymology , Mastitis, Bovine , Proto-Oncogene Proteins c-akt , Animals , Buffaloes/genetics , Buffaloes/metabolism , Cattle/genetics , Cattle/metabolism , Mastitis, Bovine/enzymology , Mastitis, Bovine/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Species Specificity
7.
Microb Pathog ; 120: 64-70, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29635052

ABSTRACT

Bacterial infection in the mammary gland parenchyma induces local and subsequently systemic inflammation that results in a complex disease. Mastitis in bovine is the result of various factors which function together. This review is aimed to analyze the factors involved in the pathogenesis of common bacterial species for bovine mastitis. The bacterial growth patterns, signaling pathway and the pathogen-associated molecular patterns (PAMPs) which activate immune responses is discussed. Clear differences in bacterial infection pattern are shown between bacterial species and illustrated TLRs, NLRs and RLGs molecular mechanism for the initiation of intramammary infection. The underlying reasons for the differences and the resulting host response are analyzed. Understandings of the mechanisms that activate and regulate these responses are central to the development of efficient anticipatory and treatment management. The knowledge of bovine mammary gland to common mastitis causing pathogens with possible immune mechanism could be a new conceptual understanding for the prospect of mastitis control program.


Subject(s)
Bacterial Infections/immunology , Host-Pathogen Interactions/immunology , Mammary Glands, Animal/immunology , Mastitis, Bovine/immunology , Receptors, Pattern Recognition/immunology , Virus Diseases/immunology , Animals , Apoptosis/immunology , Bacterial Infections/veterinary , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Cattle , Cytokines/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/veterinary , Female , Immunity, Innate/immunology , Inflammation/immunology , Mastitis, Bovine/microbiology , Mastitis, Bovine/virology , NLR Proteins/immunology , Necrosis/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Virus Diseases/veterinary
8.
Reprod Biol ; 17(4): 380-388, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29097083

ABSTRACT

Melatonin receptor 1 (MT1) performs a critical role in the regulation of the animal reproductive system, particularly in follicular growth, and has a considerable effect on reproductive performance. However, the role that MT1 plays in regulating hormones associated with reproduction remains unclear. This study was designed to examine the physiological role of constitutive MT1 silencing and follicle stimulating hormone (FSH) treatment in reproduction, making use of mouse granulosa cells (mGCs) as a model. To understand the constitutive role of MT1 in ovarian physiology, the RNAi-Ready pSIREN-RETROQ-ZsGreen Vector mediated recombinant pshRNA was used to silence MT1 gene expression. Furthermore, we observed that the expression of MT1 was successfully inhibited both at the protein and mRNA levels (P<0.001). We demonstrated that RNAi-B-mediated MT1 down-regulation significantly promoted apoptosis (P<0.001), inhibited proliferation, and regulated the cell cycle at the S-phase; conversely, FSH treatment partially aided the apoptotic effect and improved proliferation but showed a significant effect at the S-phase of the cell cycle. Transitory knockdown of MT1 proved essential in the function of mGCs, as it significantly decreased cyclic adenosine monophospahte (cAMP) level and increased cell apoptosis. Following knockdown of MT1, the expression of Bax was significantly up-regulated (P<0.001), but Bcl-2 was slightly down-regulated, both at the transcriptional and at translational levels. Moreover, the silencing of MT1 and its constitutive effect on FSH significantly promoted an increase in estradiol (P<0.001) and slightly decreased the concentration of progesterone. Together, our data indicates that MT1 suppression leads to interference in the normal physiological function of the ovary by enhancing follicular apoptosis, inhibiting proliferation, and influencing hormonal signaling, whereas constitutive FSH treatment counteracted the negative down-regulatory effects of MT1 on mGCs.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/metabolism , Receptor, Melatonin, MT1/genetics , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Estradiol/metabolism , Female , Gene Knockdown Techniques , Gene Silencing , Granulosa Cells/drug effects , Mice , Progesterone/metabolism , RNA Interference , Receptor, Melatonin, MT1/metabolism , Up-Regulation/drug effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
9.
Cell Physiol Biochem ; 44(3): 948-966, 2017.
Article in English | MEDLINE | ID: mdl-29179183

ABSTRACT

Long non-coding RNAs (lncRNAs), a class of non-coding transcripts, have recently been emerging with heterogeneous molecular actions, adding a new layer of complexity to gene-regulation networks in tumorigenesis. LncRNAs are considered important factors in several ovarian cancer histotypes, although few have been identified and characterized. Owing to their complexity and the lack of adapted molecular technology, the roles of most lncRNAs remain mysterious. Some lncRNAs have been reported to play functional roles in ovarian cancer and can be used as classifiers for personalized medicine. The intrinsic features of lncRNAs govern their various molecular mechanisms and provide a wide range of platforms to design different therapeutic strategies for treating cancer at a particular stage. Although we are only beginning to understand the functions of lncRNAs and their interactions with microRNAs (miRNAs) and proteins, the expanding literature indicates that lncRNA-miRNA interactions could be useful biomarkers and therapeutic targets for ovarian cancer. In this review, we discuss the genetic variants of lncRNAs, heterogeneous mechanisms of actions of lncRNAs in ovarian cancer tumorigenesis, and drug resistance. We also highlight the recent developments in using lncRNAs as potential prognostic and diagnostic biomarkers. Lastly, we discuss potential approaches for linking lncRNAs to future gene therapies, and highlight future directions in the field of ovarian cancer research.


Subject(s)
Ovarian Neoplasms/diagnosis , RNA, Long Noncoding/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CRISPR-Cas Systems/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Prognosis , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics
10.
Front Pharmacol ; 8: 606, 2017.
Article in English | MEDLINE | ID: mdl-28928662

ABSTRACT

In the last two decades, nanotechnologies demonstrated various applications in different fields, including detection, sensing, catalysis, electronics, and biomedical sciences. However, public concerns regarding the well-being of human may hinder the wide utilization of this promising innovation. Although, humans are exposed to airborne nanosized particles from an early age, exposure to such particles has risen dramatically within the last century due to anthropogenic sources of nanoparticles. The wide application of nanomaterials in industry, consumer products, and medicine has raised concerns regarding the potential toxicity of nanoparticles in humans. In this review, the effects of nanomaterials on the reproductive system in animal models are discussed. Females are particularly more vulnerable to nanoparticle toxicity, and toxicity in this population may affect reproductivity and fetal development. Moreover, various types of nanoparticles have negative impacts on male germ cells, fetal development, and the female reproductive system. These impacts are associated with nanoparticle modification, composition, concentration, route of administration, and the species of the animal. Therefore, understanding the impacts of nanoparticles on animal growth and reproduction is essential. Many studies have examined the effects of nanoparticles on primary and secondary target organs, with a concentration on the in vivo and in vitro effects of nanoparticles on the male and female reproductive systems at the clinical, cellular, and molecular levels. This review provides important information regarding organism safety and the potential hazards of nanoparticle use and supports the application of nanotechnologies by minimizing the adverse effects of nanoparticles in vulnerable populations.

11.
J Steroid Biochem Mol Biol ; 174: 133-140, 2017 11.
Article in English | MEDLINE | ID: mdl-28851672

ABSTRACT

Sertoli cells produce anti-Müllerian hormone (AMH), a glycoprotein belonging to the transforming growth factor-beta family. AMH mediates the regression of Müllerian ducts in the developing male fetus. However, the role of AMH in the regulation of primary Sertoli cells remains unclear. The present study was designed to investigate the effect of AMH on the viability and proliferation of Sertoli cells, with an additional focus on stem cell factor (SCF). Treatment of Sertoli cells with increasing concentrations of rh-AMH (0, 10, 50, 100, and 800ng/ml) for two days revealed that AMH, at high concentrations, increased apoptosis. These results were confirmed by a significant increase in Caspase-3 and Bax and a decrease in Bcl-2 protein and mRNA expression (P<0.01). Paradoxically, treatment with a low concentration of rh-AMH (10ng/ml), but not higher concentrations (50-800ng/ml), promoted Sertoli cell proliferation, which was verified by an increase in PCNA mRNA (P<0.05). Furthermore, only low concentrations of rh-AMH activated the non-canonical ERK signaling pathway. Similarly, low concentrations of rh-AMH (10-50ng/ml) significantly increased (P<0.05) SCF mRNA and SCF protein levels. These findings indicate that AMH differentially regulates the fate of Sertoli cells in vitro by promoting proliferation at low concentrations and apoptosis at high concentrations. In addition, AMH increased the expression of SCF, an important regulator of Sertoli cell development. Therefore, AMH may play a role in Sertoli cell development.


Subject(s)
Anti-Mullerian Hormone/metabolism , Sertoli Cells/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation , Cell Survival , Male , Mice , Stem Cell Factor
12.
Front Physiol ; 8: 354, 2017.
Article in English | MEDLINE | ID: mdl-28659810

ABSTRACT

Mature spermatozoa have highly condensed DNA that is essentially silent both transcriptionally and translationally. Therefore, post translational modifications are very important for regulating sperm motility, morphology, and for male fertility in general. Protein sumoylation was recently demonstrated in human and rodent spermatozoa, with potential consequences for sperm motility and DNA integrity. We examined the expression and localization of small ubiquitin-related modifier-1 (SUMO-1) in the sperm of water buffalo (Bubalus bubalis) using immunofluorescence analysis. We confirmed the expression of SUMO-1 in the acrosome. We further found that SUMO-1 was lost if the acrosome reaction was induced by calcium ionophore A23187. Proteins modified or conjugated by SUMO-1 in water buffalo sperm were pulled down and analyzed by mass spectrometry. Sixty proteins were identified, including proteins important for sperm morphology and motility, such as relaxin receptors and cytoskeletal proteins, including tubulin chains, actins, and dyneins. Forty-six proteins were predicted as potential sumoylation targets. The expression of SUMO-1 in the acrosome region of water buffalo sperm and the identification of potentially SUMOylated proteins important for sperm function implicates sumoylation as a crucial PTM related to sperm function.

13.
J Dairy Res ; 84(1): 76-79, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28252361

ABSTRACT

The objective of the studies presented in this Research Communication was to investigate the association of single nucleotide polymorphisms present in the MAP4K4 gene with different milk traits in dairy cows. Based on previous QTL fine mapping results on bovine chromosome 11, the MAP4K4 gene was selected as a candidate gene to evaluate its effect on somatic cell count and milk traits in ChineseHolstein cows. Milk production traits including milk yield, fat percentage, and protein percentage of each cow were collected using 305 d lactation records. Association between MAP4K4 genotype and different traits and Somatic Cell Score (SCS) was performed using General Linear Regression Model of R. Two SNPs at exon 18 (c.2061T > G and c.2196T > C) with genotype TT in both SNPs were found significantly higher for somatic SCS. We found the significant effect of exon 18 (c.2061T > G) on protein percentage, milk yield and SCS. We identified SNPs at different location of MAP4K4 gene of the cattle and several of them were significantly associated with the somatic cell score and other different milk traits. Thus, MAP4K4 gene could be a useful candidate gene for selection of dairy cattle against mastitis and the identified polymorphisms might potentially be strong genetic markers.


Subject(s)
Lactation/genetics , Mastitis, Bovine/genetics , Milk/chemistry , Polymorphism, Single Nucleotide/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Cattle , Cell Count , China , Chromosome Mapping , Exons/genetics , Fats/analysis , Female , Genetic Markers , Linear Models , Mastitis, Bovine/enzymology , Milk/cytology , Milk Proteins/analysis , Quantitative Trait Loci
14.
Int J Mol Sci ; 18(2)2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28208755

ABSTRACT

Our understanding of the post-transcriptional mechanisms involved in follicular atresia is limited; however, an important development has been made in understanding the biological regulatory networks responsible for mediating follicular atresia. MicroRNAs have come to be seen as a key regulatory actor in determining cell fate in a wide range of tissues in normal and pathological processes. Profiling studies of miRNAs during follicular atresia and development have identified several putative miRNAs enriched in apoptosis signaling pathways. Subsequent in vitro and/or in vivo studies of granulosa cells have elucidated the functional role of some miRNAs along with their molecular pathways. In particular, the regulatory roles of some miRNAs have been consistently observed during studies of follicular cellular apoptosis. Continued work should gradually lead to better understanding of the role of miRNAs in this field. Ultimately, we expect this understanding will have substantial benefits for fertility management at both the in vivo or/and in vitro levels. The stable nature of miRNA holds remarkable promise in clinical use as a diagnostic tool and in reproductive medicine to solve the ever-increasing fertility problem. In this review, we summarize current knowledge of the involvement of miRNAs in follicular atresia, discuss the challenges for further work and pinpoint areas for future research.


Subject(s)
Follicular Atresia/genetics , Gene Expression Regulation , MicroRNAs/genetics , Animals , Apoptosis/genetics , Autophagy/genetics , Biomarkers , Female , Follicular Atresia/metabolism , Gene Expression Profiling , Granulosa Cells/metabolism , Humans , Ovarian Follicle/metabolism , Signal Transduction , Transcriptome
15.
Anim Reprod Sci ; 172: 76-82, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27449408

ABSTRACT

The aim of this study was to evaluate the efficacy of an inhibin DNA vaccine delivered by attenuated Salmonella choleraesuis on follicular development and ovulation responses in crossbred buffaloes. A total of 158 crossbred buffaloes divided into four groups and were intramuscularly injected with 1×10(10) (T1, n=41), 1×10(9) (T2, n=37), 1×10(8) (T3, n=37) or 0 (C, n=43) CFU/ml bacteria delivered inhibin vaccine in 10ml PBS on day 0 and 14, respectively. All animals were administered with 1000 IU PMSG on day 28, 0.5mg PGF2α on day 30 and 200µg GnRH on day 32. The results showed buffaloes immunized with the bacteria delivered inhibin vaccine had significantly higher titers of anti-inhibin IgG antibody than control group (P<0.01). The number and diameter of large follicles (≥10mm) as well as ovulatory follicles in group T1 was significantly greater than group C (P<0.05). The growth speed of dominant follicles in group T1 was significantly faster than groups T3 and C (P<0.05), resulting in a greater conception rate in buffaloes with positive antibodies. These results demonstrate that immunization with the bacterial delivered inhibin vaccine, coupled with the estrus synchronization protocol, could be used as an alternative approach to improve fertility in crossbred buffaloes.


Subject(s)
Buffaloes/physiology , Inhibins/immunology , Ovulation/physiology , Animals , Estrus Synchronization , Female , Ovarian Follicle , Salmonella/genetics , Vaccines, Attenuated/immunology , Vaccines, DNA/immunology , Vaccines, Synthetic
16.
J Steroid Biochem Mol Biol ; 152: 161-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25998417

ABSTRACT

Inhibin α (INHα), a member of TGFß superfamily, is an important modulator of reproductive function that plays a vital role in follicular changes, cell differentiation, oocyte development, and ultimately in mammalian reproduction. However, the role of inhibin α in female fertility and ovarian function remains largely unknown. To define its role in reproduction, transgenic mice of RNAi-INHα that knock down the INHα expression by shRNAi were used. Inhibin α subunit gene was knocked down successfully at both transcriptional and translational levels by RNAi PiggyBac transposon (Pbi) mediated recombinant pshRNA vectors and purified DNA fragments were microinjected into mouse zygotes. Results showed that transgenic female mice were sub-fertile and exhibited 35.28% reduction in litter size in F1 generation relative to wild type. The decreased litter size associated with the reduction in the number of oocytes ovulated after puberty. Serum INHα level was significantly decreased in both 3 and 6 weeks; whereas, FSH was significantly increased in 3 weeks but not in 6 weeks. Furthermore, suppression of INHα expression significantly promoted apoptosis by up-regulating Caspase-3, bcl2, INHßB and GDF9 and down regulated Kitl and TGFßRIII genes both at transcriptional and translational levels. Moreover, it also dramatically reduced the progression of G1 phase of cell cycle and the number of cells in S phase as determined by flow cytometer. These results indicate that suppression of INHα expression in RNAi-transgenic mice leads to disruption of normal ovarian regulatory mechanism and causes reproductive deficiencies by promoting cellular apoptosis, arresting cellular progression and altering hormonal signaling.


Subject(s)
Apoptosis/genetics , Fertility/genetics , Granulosa Cells/metabolism , Inhibins/genetics , Ovulation/genetics , Animals , Female , Follicle Stimulating Hormone/blood , G1 Phase Cell Cycle Checkpoints/genetics , Inhibins/blood , Litter Size/genetics , Luteinizing Hormone/blood , Mice , Mice, Transgenic , Oocytes/cytology , Puberty, Delayed/genetics , RNA Interference , RNA, Small Interfering
SELECTION OF CITATIONS
SEARCH DETAIL
...