Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546420

ABSTRACT

Members of the Tribbles (TRIB) family of pseudokinases are critical components of intracellular signal transduction pathways in physiological and pathological processes. TRIBs, including TRIB2, have been previously shown as signaling mediators and scaffolding proteins regulating numerous cellular events such as proliferation, differentiation and cell death through protein stability and activity. However, the signaling network associated with TRIB2 and its binding partners in granulosa cells during ovarian follicular development is not fully defined. We previously reported that TRIB2 is differentially expressed in growing dominant follicles while downregulated in ovulatory follicles following the luteinizing hormone (LH) surge or human chorionic gonadotropin (hCG) injection. In the present study, we used the yeast two-hybrid screening system and in vitro coimmunoprecipitation assays to identify and confirm TRIB2 interactions in granulosa cells (GCs) of dominant ovarian follicles (DFs), which yielded individual candidate binding partners including calmodulin 1 (CALM1), inhibin subunit beta A (INHBA), inositol polyphosphate phosphatase-like 1 (INPPL1), 5'-nucleotidase ecto (NT5E), stearoyl-CoA desaturase (SCD), succinate dehydrogenase complex iron sulfur subunit B (SDHB) and Ras-associated protein 14 (RAB14). Further analyses showed that all TRIB2 binding partners are expressed in GCs of dominant follicles but are differentially regulated throughout the different stages of follicular development. CRISPR/Cas9-driven inhibition along with pQE-driven overexpression of TRIB2 showed that TRIB2 differently regulates expression of binding partners, which reveals the importance of TRIB2 in the control of gene expression linked to various biological processes such as proliferation, differentiation, cell migration, apoptosis, calcium signaling and metabolism. These data provide a larger view of potential TRIB2-regulated signal transduction pathways in GCs and provide strong evidence that TRIB2 may act as a regulator of target genes during ovarian follicular development.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Regulation , Granulosa Cells/metabolism , Animals , Biomarkers , Cattle , Down-Regulation , Female , Ovarian Follicle/metabolism , Protein Binding , Protein Interaction Mapping , Two-Hybrid System Techniques
2.
PLoS One ; 14(2): e0212571, 2019.
Article in English | MEDLINE | ID: mdl-30811458

ABSTRACT

Ankyrin-repeat and SOCS-box protein 9 (ASB9) is a member of the large SOCS-box containing proteins family and acts as the specific substrate recognition component of E3 ubiquitin ligases in the process of ubiquitination and proteasomal degradation. We previously identified ASB9 as a differentially expressed gene in granulosa cells (GC) of bovine ovulatory follicles. This study aimed to further investigate ASB9 mRNA and protein regulation, identify binding partners in GC of bovine ovulatory follicles, and study its function. GC were obtained from small follicles (SF: 2-4 mm), dominant follicles at day 5 of the estrous cycle (DF), and ovulatory follicles, 24 hours following hCG injection (OF). Analyses by RT-PCR showed a 104-fold greater expression of ASB9 in GC of OF than in DF. Steady-state levels of ASB9 in follicular walls (granulosa and theca cells) analyzed at 0, 6, 12, 18 and 24 hours after hCG injection showed a significant induction of ASB9 expression at 12 and 18 hours, reaching a maximum induction of 10.2-fold at 24 hours post-hCG as compared to 0 hour. These results were confirmed in western blot analysis showing strongest ASB9 protein amounts in OF. Yeast two-hybrid screening of OF-cDNAs library resulted in the identification of 10 potential ASB9 binding partners in GC but no interaction was found between ASB9 and creatine kinase B (CKB) in these GC. Functional studies using CRISPR-Cas9 approach revealed that ASB9 inhibition led to increased GC proliferation and modulation of target genes expression. Overall, these results support a physiologically relevant role of ASB9 in the ovulatory follicle by targeting specific proteins likely for degradation, contributing to reduced GC proliferation, and could be involved in the final GC differentiation into luteal cells.


Subject(s)
Chorionic Gonadotropin/metabolism , Gene Expression Regulation, Developmental , Granulosa Cells/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Theca Cells/metabolism , Animals , Ankyrin Repeat , Cattle , Cell Differentiation , Cell Proliferation , Chorionic Gonadotropin/administration & dosage , Female , Luteal Cells/physiology , Models, Animal , Protein Binding , Protein Interaction Mapping , Proteolysis , Suppressor of Cytokine Signaling Proteins/genetics
3.
Reprod Biol Endocrinol ; 15(1): 88, 2017 Nov 03.
Article in English | MEDLINE | ID: mdl-29100496

ABSTRACT

BACKGROUND: Ovulation and luteinization of follicles are complex biological processes initiated by the preovulatory luteinizing hormone surge. The objective of this study was to identify genes that are differentially expressed in bovine granulosa cells (GC) of ovulatory follicles. METHODS: Granulosa cells were collected during the first follicular wave of the bovine estrous cycle from dominant follicles (DF) and from ovulatory follicles (OF) obtained 24 h following injection of human chorionic gonadotropin (hCG). A granulosa cell subtracted cDNA library (OF-DF) was generated using suppression subtractive hybridization and screened. RESULTS: Detection of genes known to be upregulated in bovine GC during ovulation, such as ADAMTS1, CAV1, EGR1, MMP1, PLAT, PLA2G4A, PTGES, PTGS2, RGS2, TIMP1, TNFAIP6 and VNN2 validated the physiological model and analytical techniques used. For a subset of genes that were identified for the first time, gene expression profiles were further compared by semiquantitative RT-PCR in follicles obtained at different developmental stages. Results confirmed an induction or upregulation of the respective mRNAs in GC of OF 24 h after hCG-injection compared with those of DF for the following genes: ADAMTS9, ARAF, CAPN2, CRISPLD2, FKBP5, GFPT2, KIT, KITLG, L3MBLT3, MRO, NUDT10, NUDT11, P4HA3, POSTN, PSAP, RBP1, SAT1, SDC4, TIMP2, TNC and USP53. In bovine GC, CRISPLD2 and POSTN mRNA were found as full-length transcript whereas L3MBLT3 mRNA was alternatively spliced resulting in a truncated protein missing the carboxy-terminal end amino acids, 774KNSHNEL780. Conversely, L3MBLT3 is expressed as a full-length mRNA in a bovine endometrial cell line. The 774KNSHNEL780 sequence is well conserved in all mammalian species and follows a SAM domain known to confer protein/protein interactions, which suggest a key function for these amino acids in the epigenetic control of gene expression. CONCLUSIONS: We conclude that we have identified novel genes that are upregulated by hCG in bovine GC of OF, thereby providing novel insight into peri-ovulatory regulation of genes that contribute to ovulation and/or luteinization processes.


Subject(s)
Chorionic Gonadotropin/pharmacology , Granulosa Cells/drug effects , Ovarian Follicle/drug effects , RNA, Messenger/metabolism , Up-Regulation/drug effects , Animals , Cattle , Female , Gene Expression/drug effects , Gene Expression Profiling , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , Ovulation/drug effects , RNA, Messenger/genetics
4.
J Ovarian Res ; 9(1): 71, 2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27793176

ABSTRACT

BACKGROUND: Janus kinase 3 (JAK3) is a member of the membrane-associated non-receptor tyrosine kinase protein family and is considered predominantly expressed in hematopoietic cells. We previously identified JAK3 as a differentially expressed gene in granulosa cells (GC) of bovine preovulatory follicles. The present study aimed to further investigate JAK3 regulation, to identify protein binding partners and better understand its mode of action in bovine reproductive cells. RESULTS: GC were obtained from small follicles (SF), dominant follicles at day 5 of the estrous cycle (DF), and ovulatory follicles, 24 h following hCG injection (OF). RT-PCR analyses showed greatest expression of JAK3 in GC of DF, while JAK3 expression was downregulated in OF (P < 0.0001). In addition, there was a 5- and 20-fold reduction of JAK3 steady-state mRNA levels in follicular walls, respectively at 12 and 24 hours post-hCG as compared to 0 h (P < 0.05). Similarly, JAK3 expression was downregulated by the endogenous LH surge. These results were confirmed in western blot analysis showing weakest JAK3 protein amounts in OF as compared to DF. Yeast two-hybrid screening of a DF-cDNA library resulted in the identification of JAK3 partners in GC that were confirmed by co-immunoprecipitation and included leptin receptor overlapping transcript-like 1 (LEPROTL1), inhibin beta A (INHBA) and cyclin-dependent kinase inhibitor 1B (CDKN1B). In functional studies using bovine endometrial cells, JAK3 increased phosphorylation of STAT3 and cell viability, while the addition of JANEX-1 inhibited JAK3 actions. CONCLUSION: These results support a physiologically relevant role of JAK3 in follicular development and provide insights into the mode of action and function of JAK3 in reproductive tissues.


Subject(s)
Carrier Proteins/metabolism , Granulosa Cells/metabolism , Janus Kinase 3/metabolism , Ovarian Follicle/metabolism , Ovulation/metabolism , Animals , Cattle , Cell Survival/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endometrium/metabolism , Female , Gene Expression Regulation , Inhibin-beta Subunits/metabolism , Janus Kinase 3/genetics , Ovulation/genetics , Protein Binding , Protein Interaction Mapping , RNA, Messenger/genetics , STAT Transcription Factors/metabolism
5.
J Ovarian Res ; 8: 12, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25881887

ABSTRACT

BACKGROUND: LAPTM4B is a member of the lysosome-associated transmembrane protein superfamily that is differentially expressed in normal human tissues and upregulated in various types of carcinomas. These proteins are thought to be involved in the regulation of cell proliferation and survival. The objective of this study was to investigate the expression of bovine LAPTM4B during ovarian follicular development and in various bovine tissues. METHODS AND RESULTS: Northern blot analysis revealed a 1.8 kb transcript, with highly variable steady state levels among tissues. RT-PCR analysis showed that LAPTM4B mRNA transcripts were low in granulosa cells of small antral follicles, increased in large dominant follicles, and decreased in ovulatory follicles following injection of human chorionic gonadotropin (hCG; P < 0.003). Ovulatory follicles collected at various times after hCG injection revealed a significant reduction of LAPTM4B mRNA starting at 18 h post-hCG (P < 0.029). Immunoblotting analysis using antibodies generated against bovine LAPTM4B recognized proteins of 26.3 and 31.5 kDa in granulosa cells of developing follicles and corpus luteum. Further analyses of affinity-purified His-tag LAPTM4B overexpressed in HEK cells showed that the 31.5 kDa protein represented the ubiquinated isoform of the 26.3 kDa native protein. The 26.3 kDa protein was differentially expressed showing highest amounts in dominant follicles and lowest amounts in ovulatory follicles 24 h post-hCG. Immunohistochemical analyses of LAPTM4B showed marked heterogeneity of labeling signal among tissues, with LAPTM4B mainly localized to perinuclear vesicles, in keeping with its putative lysosomal membrane localization. CONCLUSION: This study reports for the first time that bovine LAPTM4B in granulosa cells is present in both unubiquinated and ubiquinated forms, and is differentially expressed in developing ovarian follicles, suggesting a possible role in terminal follicular growth.


Subject(s)
Granulosa Cells/metabolism , Membrane Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Animals , Blotting, Northern , Cattle , Female , Immunohistochemistry , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Ubiquitination
6.
Biol Reprod ; 91(6): 139, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25339105

ABSTRACT

The luteinizing hormone preovulatory surge stimulates several signal pathways essential for ovulation, and the regulator of G-protein signaling protein-2 (RGS2) is thought to be involved in this process. The objectives of this study were to characterize the regulation of RGS2 transcripts in equine and bovine follicles prior to ovulation and to determine its transcriptional control in bovine granulosa cells. To assess the regulation of equine RGS2 prior to ovulation, RT-PCR was performed using total RNA extracted from equine follicles collected at various times after human chorionic gonadotropin (hCG) injection. Results showed that RGS2 mRNA levels were very low at 0 h but markedly increased 12-39 h post-hCG (P < 0.05). In the bovine species, results revealed that RGS2 mRNA levels were low in small and dominant follicles and in ovulatory follicles obtained at 0 h, but markedly increased in ovulatory follicles 6-24 h post-hCG (P < 0.05). To study the molecular control of RGS2 expression, primary cultures of bovine granulosa cells were used. Stimulation with forskolin induced an up-regulation of RGS2 mRNA in vitro. Studies using 5'-deletion mutants identified a minimal region containing full-length basal and forskolin-inducible RGS2 promoter activities. Site-directed mutagenesis indicated that these activities were dependent on CRE and ETS1 cis-elements. Electrophoretic mobility shift assays confirmed the involvement of these elements and revealed their interactions with CREB1 and ETS1 proteins. Chromatin immunoprecipitation assays confirmed endogenous interactions of these proteins with the RGS2 promoter in granulosa cells. Forskolin-inducible RGS2 promoter activity and mRNA expression were markedly decreased by PKA and ERK1/2 inhibitors, and treatment with an antagonist of PGR (RU486) and inhibitors of PTGS2 (NS398) and EGFR (PD153035) blocked the forskolin-dependent RGS2 transcript expression, suggesting the importance of RGS2 in ovulation. Collectively, this study reports for the first time the gonadotropin-dependent up-regulation of RGS2 in equine and bovine preovulatory follicles and presents some of the regulatory controls involved in RGS2 gene expression in granulosa cells.


Subject(s)
Cattle/genetics , Granulosa Cells/metabolism , Horses/genetics , Ovarian Follicle/metabolism , Ovulation/genetics , RGS Proteins/genetics , Animals , Cattle/physiology , Cells, Cultured , Female , Follicular Phase/drug effects , Follicular Phase/genetics , Follicular Phase/metabolism , Gene Expression Regulation/drug effects , Gonadotropins/pharmacology , Granulosa Cells/drug effects , Horses/physiology , Ovarian Follicle/drug effects , Ovulation/drug effects , Ovulation/metabolism , RGS Proteins/metabolism , Transcriptional Activation/drug effects
7.
Biol Reprod ; 89(4): 98, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24006283

ABSTRACT

Vanin-2 (VNN2) is known to be involved in inflammation and leukocyte migration, but its regulation in follicles remains unknown. The objectives of this work were to study the regulation of VNN2 transcripts in bovine follicles prior to ovulation and to characterize the control of its expression in bovine granulosa cells. VNN2 expression was studied using total RNA extracted from granulosa cells of small follicles (2-4 mm in diameter), dominant follicles obtained on Day 5 of the estrous cycle, ovulatory follicles obtained 0-24 h after human chorionic gonadotropin (hCG), and corpora lutea on Day 5 of the cycle. The results from RT-PCR analyses showed that levels of VNN2 mRNA were high in ovulatory follicles 24 h post-hCG but low in the other tissues. In ovulatory follicles, levels of VNN2 mRNA were low at 0 h but significantly up-regulated 12-24 h post-hCG. To determine factors controlling VNN2 gene expression, established primary cultures of granulosa cells isolated from bovine dominant follicles were used. Treatment with forskolin elevated VNN2 mRNA expression as observed in vivo. Mutation studies identified the minimal region conferring basal and forskolin-stimulated VNN2 promoter activities, which were dependent on chicken ovalbumin upstream promoter-transcription factor (COUP-TF), GATA, and Ebox cis-elements. Electrophoretic mobility shift assays identified COUP-TF, GATA4, and upstream stimulating factor proteins as key factors interacting with these elements. Chromatin immunoprecipitation assays confirmed basal and forskolin-induced interactions between these proteins and the VNN2 promoter in bovine granulosa cell cultures. VNN2 promoter activity and mRNA expression were markedly stimulated by forskolin and overexpression of the catalytic subunit of PKA, but inhibited by PKA and ERK1/2 inhibitors. Collectively, the findings from this study describe for the first time the gonadotropin/forskolin-dependent up-regulation of VNN2 transcripts in granulosa cells of preovulatory follicles and provide insights into some of the molecular bases of VNN2 gene expression in follicular cells.


Subject(s)
Amidohydrolases/metabolism , Cell Adhesion Molecules/metabolism , Ovarian Follicle/metabolism , Proestrus , Promoter Regions, Genetic , Transcription, Genetic , Up-Regulation , Amidohydrolases/biosynthesis , Amidohydrolases/genetics , Animals , Cattle , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Colforsin/pharmacology , Corpus Luteum/cytology , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Enzyme Activators/pharmacology , Female , Fertility Agents, Female/pharmacology , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Granulosa Cells/cytology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mutation , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Proestrus/drug effects , Promoter Regions, Genetic/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Response Elements/drug effects , Transcription, Genetic/drug effects , Up-Regulation/drug effects
8.
Mol Cancer ; 9: 271, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20942929

ABSTRACT

BACKGROUND: Among the most harmful of all genetic abnormalities that appear in colorectal cancer (CRC) development are mutations of KRAS and its downstream effector BRAF as they result in abnormal extracellular signal-related kinase (ERK) signaling. In a previous report, we had shown that expression of a constitutive active mutant of MEK1 (caMEK) in normal rat intestinal epithelial cells (IECs) induced morphological transformation associated with epithelial to mesenchymal transition, growth in soft agar, invasion and metastases in nude mice. Results from microarrays comparing control to caMEK-expressing IECs identified the gene encoding for serpinE2, a serine protease inhibitor, as a potential target of activated MEK1. RESULTS: 1- RT-PCR and western blot analyses confirmed the strong up-regulation of serpinE2 expression and secretion by IECs expressing oncogenic MEK, Ras or BRAF. 2- Interestingly, serpinE2 mRNA and protein were also markedly enhanced in human CRC cells exhibiting mutation in KRAS and BRAF. 3- RNAi directed against serpinE2 in caMEK-transformed rat IECs or in human CRC cell lines HCT116 and LoVo markedly decreased foci formation, anchorage-independent growth in soft agarose, cell migration and tumor formation in nude mice. 4- Treatment of CRC cell lines with U0126 markedly reduced serpinE2 mRNA levels, indicating that expression of serpinE2 is likely dependent of ERK activity. 5- Finally, Q-PCR analyses demonstrated that mRNA levels of serpinE2 were markedly increased in human adenomas in comparison to healthy adjacent tissues and in colorectal tumors, regardless of tumor stage and grade. CONCLUSIONS: Our data indicate that serpinE2 is up-regulated by oncogenic activation of Ras, BRAF and MEK1 and contributes to pro-neoplastic actions of ERK signaling in intestinal epithelial cells. Hence, serpinE2 may be a potential therapeutic target for colorectal cancer treatment.


Subject(s)
Colorectal Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Serpin E2/metabolism , Animals , Blotting, Western , Butadienes/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , HCT116 Cells , Humans , In Vitro Techniques , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mice , Mice, Nude , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction , Serpin E2/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , ras Proteins/genetics , ras Proteins/metabolism
9.
Reprod Biol Endocrinol ; 8: 94, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20682045

ABSTRACT

The endoplasmic reticulum (ER) is a major site of protein synthesis and facilitates the folding and assembly of newly synthesized proteins. Misfolded proteins are retrotranslocated across the ER membrane and destroyed at the proteasome. DERL1 is an important protein involved in the retrotranslocation and degradation of a subset of misfolded proteins from the ER. We characterized a 2617 bp cDNA from bovine granulosa cells that corresponded to bovine DERL1. Two transcripts of 3 and 2.6 kb were detected by Northern blot analysis, and showed variations in expression among tissues. During follicular development, DERL1 expression was greater in day 5 dominant follicles compared to small follicles, ovulatory follicles, or corpus luteum (CL). Within the CL, DERL1 mRNA expression was intermediate in midcycle, and lowest in late cycle as compared to early in the estrous cycle. Western blot analyses demonstrated the presence of DERL1 in the bovine CL at days 5, 11, and 18 of the estrous cycle. Co-immunoprecipitation using luteal tissues showed that DERL1 interacts with class I MHC but not with VIMP or p97 ATPase. The interaction between DERL1 and MHC I suggests that, in the CL, DERL1 may regulate the integrity of MHC I molecules that are transported to the ER membrane. Furthermore, the greater expression of DERL1 mRNA is associated with the active follicular development and early luteal stages, suggesting a role of DERL1 in tissue remodeling events and maintenance of function in reproductive tissues.


Subject(s)
Cattle/genetics , Corpus Luteum/metabolism , Membrane Proteins/genetics , Ovarian Follicle/metabolism , Animals , Cattle/metabolism , Cloning, Molecular , Female , Gene Expression , Gene Expression Profiling , Membrane Proteins/metabolism , Membrane Proteins/physiology , Ovarian Follicle/physiology , RNA, Messenger/metabolism , Tissue Distribution
10.
Physiol Genomics ; 38(2): 149-57, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19401403

ABSTRACT

Wound healing in horses is complicated, particularly when wounds are on the limb. The objectives of this study were to clone equine thrombospondin II (THBS2) and secreted protein acidic and cysteine-rich (SPARC) cDNAs and to compare the spatiotemporal expression of mRNAs and proteins during repair of body and limb wounds. These molecules were targeted in view of their potential biological contribution to angiogenesis, which is exacerbated during the repair of limb wounds in horses. Cloning was achieved by screening size-selected cDNA libraries previously derived from 7-day-old wounds. Expression was studied in unwounded skin and in samples from 1, 2, 3, 4, and 6 wk old wounds of the body and limb. Temporal gene expression was determined by semiquantitative RT-PCR, while protein expression was mapped immunohistochemically. The temporal pattern of expression for both genes was similar; wounding caused immediate upregulation of mRNA, which did not return to baseline by the end of the study, and overexpression was noted in body relative to limb wounds. Immunostaining for THBS2 and SPARC was induced by wounding, though no differences in stain location or intensity were detected between body and limb wounds. This study is the first to characterize equine cDNA for THBS2 and SPARC and to document mRNA expression over the different phases of repair. THBS2 and SPARC might modulate angiogenesis during wound healing in the horse, which could protect against the disproportionate fibroplasia commonly afflicting limb wounds and leading to the development of exuberant granulation tissue.


Subject(s)
Cell Adhesion Molecules/metabolism , Osteonectin/metabolism , RNA, Messenger/metabolism , Thrombospondins/metabolism , Wound Healing/physiology , Animals , Cell Adhesion Molecules/genetics , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , Horses , Immunoblotting , Immunohistochemistry , Linear Models , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Osteonectin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondins/genetics
11.
Vet Dermatol ; 20(4): 243-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19374723

ABSTRACT

The development of exuberant granulation tissue, a situation that in some ways resembles the human keloid, compromises both the aesthetic and functional outcomes of wound repair in horses. To help elucidate the underlying molecular mechanisms the spatio-temporal expression of lumican (LUM) mRNA and protein for their potential contributions to tissue remodelling of body and limb wounds, was examined in an established experimental model. Expression was studied in intact skin and in samples of 1-, 2-, 3-, 4- and 6-week-old wounds of the body and forelimb. Temporal gene expression was determined by reverse transcriptase polymerase chain reaction, and protein expression was mapped immunohistochemically. A significant increase in LUM mRNA expression was observed in response to wounding at both anatomical locations, and a significantly higher mRNA level was recorded in thoracic than in limb wounds at weeks 1, 3 and 6 of repair. The immunohistochemical observations partially corroborated the mRNA data. To the authors' knowledge this study is the first to document that the cDNA for LUM is expressed over the different phases of wound repair in horses and suggests that LUM might be involved in both inflammation and remodelling in response to dermal injury. Further studies are now required to verify and quantify the temporal expression of this protein to provide the basis for targeted therapies that might prevent the development of exuberant granulation tissue in horse wound repair.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Gene Expression Regulation/physiology , Granulation Tissue/growth & development , Horses/injuries , Keratan Sulfate/metabolism , Wound Healing/physiology , Animals , Chondroitin Sulfate Proteoglycans/genetics , Cloning, Molecular , DNA, Complementary/genetics , Immunohistochemistry , Keratan Sulfate/genetics , Lumican , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin/metabolism , Wounds and Injuries/metabolism
12.
Am J Vet Res ; 70(1): 112-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19119956

ABSTRACT

OBJECTIVE: To clone full-length equine pigment epithelium-derived factor (PEDF) complementary DNA (cDNA) and to evaluate its temporal expression during repair of wounds in horses. ANIMALS: 4 clinically normal 2-to 3-year-old Standardbred mares. PROCEDURES: Full-length equine PEDF cDNA was cloned by screening size-selected cDNA libraries derived from biopsy specimens obtained from the wound edge 7 days after experimental creation of a 6.25-cm(2) full-thickness wound in the skin of the lateral thoracic wall. Expression was evaluated in normal skin and in biopsy specimens obtained weekly from experimentally induced wounds on the trunk and limbs of horses. Temporal gene expression was determined by use of reverse transcriptase PCR assay. RESULTS: Equine PEDF shared 87% sequence and 88% peptide homology with human PEDF. Wounding caused upregulation of PEDF mRNA, which did not return to baseline by the end of the study in either anatomic location. Relative overexpression was evident in wounds on the trunk, compared with expression for wounds on the limbs. CONCLUSIONS AND CLINICAL RELEVANCE: This study characterized full-length equine cDNA for PEDF and determined that the gene for PEDF appeared to be upregulated in response to dermal wounding. Although the cause of exuberant granulation tissue is probably multifactorial, these data suggested that PEDF, via its potent antiangiogenic capabilities, may contribute to superior healing in wounds on the trunks of horses by protecting such wounds from excessive formation of vascular granulation tissue that characterizes wounds on the limbs of this species.


Subject(s)
Eye Proteins/genetics , Horses/genetics , Nerve Growth Factors/genetics , Serpins/genetics , Wound Healing/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Eye Proteins/biosynthesis , Eye Proteins/blood , Female , Gene Expression , Horses/blood , Molecular Sequence Data , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/blood , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Alignment , Serpins/biosynthesis , Serpins/blood
13.
Vet Dermatol ; 20(1): 27-34, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19121151

ABSTRACT

Healing of wounds located on the distal limbs of horses is often complicated by retarded epithelialization and the development of exuberant granulation tissue (proud flesh). Treatments that definitively resolve this pathological process are still unavailable. Molecular studies of the repair mechanism might contribute to the development of new therapeutic strategies. The study presented herein aimed to clone the full length cDNA and to study the spatio-temporal expression profile of mRNA and protein for LAMR1, previously attributed a role in wound epithelialization, during the repair of body and limb wounds in the horse. Cloning was achieved by screening a cDNA library previously derived from 7-day wound biopsies. Expression was studied in unwounded skin and in samples from 1-, 2-, 3-, 4- and 6-week-old wounds of the body and limb. Temporal gene expression was determined by real time polymerase chain reaction (RT-PCR) while protein expression was mapped immunohistochemically. Full-length cDNA for equine LAMR1 was shown to be highly similar to that of other species. The mRNA expression of LAMR1 was significantly up-regulated only in thoracic wounds, 4 and 6 weeks following wounding (upon epithelialization). Cutaneous wounding induced protein expression at both locations. Our data suggest that up-regulation of LAMR1 protein might favour epithelialization during wound healing. However, its interaction with ligands other than laminin complicates data interpretation. Future studies should quantitatively verify the temporal expression of this protein in order to provide the basis for targeted therapies that might enhance epithelialization.


Subject(s)
Gene Expression Regulation/physiology , Horses/metabolism , Receptors, Laminin/metabolism , Wound Healing/physiology , Wounds and Injuries/veterinary , Animals , DNA, Complementary/genetics , Immunoblotting , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Laminin/genetics , Skin/metabolism , Wounds and Injuries/metabolism
14.
Reproduction ; 136(4): 447-57, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18635743

ABSTRACT

Survival and inhibitory factors regulate steroidogenesis and determine the fate of developing follicles. The objective of this study was to determine the role of transforming growth factor-beta1 (TGFB1) in the regulation of estradiol-17beta (E(2)) and progesterone (P(4)) secretion in FSH-stimulated bovine granulosa cells. Granulosa cells were obtained from 2 to 5 mm follicles and cultured in serum-free medium. FSH dose (1 and 10 ng/ml for 6 days) and time in culture (2, 4, and 6 days with 1 ng/ml FSH) increased E(2) secretion and mRNA expression of E(2)-related enzymes cytochrome P450 aromatase (CYP19A1) and 17beta-hydroxysteroid dehydrogenase type 1 (HSD17B1), but not HSD17B7. TGFB1 in the presence of FSH (1 ng/ml) inhibited E(2) secretion, and decreased mRNA expression of FSH receptor (FSHR), CYP19A1, and HSD17B1, but not HSD17B7. FSH dose did not affect P(4) secretion and mRNA expression of 3beta-hydroxysteroid dehydrogenase (HSD3B) and alpha-glutathione S-transferase (GSTA), but inhibited the amount of steroidogenic acute regulatory protein (STAR) mRNA. Conversely, P(4) and mRNA expression of STAR, cytochrome P450 side-chain cleavage (CYP11A1), HSD3B, and GSTA increased with time in culture. TGFB1 inhibited P(4) secretion and decreased mRNA expression of STAR, CYP11A1, HSD3B, and GSTA. TGFB1 modified the formation of granulosa cell clumps and reduced total cell protein. Finally, TGFB1 decreased conversion of androgens to E(2), but did not decrease the conversion of estrone (E(1)) to E(2) and pregnenolone to P(4). Overall, these results indicate that TGFB1 counteracts stimulation of E(2) and P(4) synthesis in granulosa cells by inhibiting key enzymes involved in the conversion of androgens to E(2) and cholesterol to P(4) without shutting down HSD17B reducing activity and HSD3B activity.


Subject(s)
Estradiol/biosynthesis , Granulosa Cells/enzymology , Progesterone/biosynthesis , Transforming Growth Factor beta1/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , Androgens/metabolism , Animals , Aromatase/genetics , Aromatase/metabolism , Cattle , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Estradiol/metabolism , Estrone/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression/drug effects , Glutathione Transferase/genetics , Phosphoproteins/genetics , Pregnenolone/metabolism , Progesterone/metabolism , RNA, Messenger/analysis , Receptors, FSH/genetics , Stimulation, Chemical , Transforming Growth Factor beta1/metabolism
15.
J Dermatol Sci ; 51(2): 103-12, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18434095

ABSTRACT

BACKGROUND: Wounds on horse limbs can develop exuberant granulation tissue which resembles the human keloid. Clues gained from the study of over-scarring in horses might help control fibro-proliferative disorders. OBJECTIVES: The aim of the present study was to clone full-length equine ANXA2 cDNA then to study spatio-temporal expression of ANXA2 and MMP1 mRNA and protein, potential contributors to remodeling, during repair of body (normal) and limb (fibro-proliferative) wounds in an established horse wound model. METHODS: Cloning of ANXA2 was achieved by screening size-selected cDNA libraries. Expression was studied in intact skin and in biopsies of 1, 2, 3, 4 and 6-week-old wounds of the body and limb. Temporal gene expression was determined by semi-quantitative RT-PCR while protein expression was mapped immunohistochemically. RESULTS: ANXA2 mRNA was up-regulated only in body wounds, corroborating the superior and prompt tissue turnover at this location. Immunohistochemistry partially substantiated the mRNA data in that increased staining for ANXA2 protein was detected in neo-epidermis which formed more rapidly and completely in body wounds. MMP1 mRNA levels in body wounds significantly surpassed those of limb wounds in week one biopsies. The protein was abundant in migrating epithelium of limb wounds at weeks two and four; conversely, body wounds in which epithelialization was near complete showed diminished staining of MMP1. CONCLUSION: We conclude that ANXA2 and MMP1 might participate in remodeling during wound healing in the horse, and that differences in expression may contribute to the excessive proliferative response seen in the limb.


Subject(s)
Annexin A2/metabolism , Matrix Metalloproteinase 1/metabolism , Skin/metabolism , Wound Healing/physiology , Wounds and Injuries/metabolism , Animals , Annexin A2/genetics , Biopsy , Cell Proliferation , DNA, Complementary/genetics , Female , Gene Expression Regulation , Horses , Matrix Metalloproteinase 1/genetics , Models, Animal , RNA, Messenger/metabolism , Skin/cytology , Skin/pathology , Wounds and Injuries/pathology
16.
BMC Physiol ; 8: 1, 2008 Jan 31.
Article in English | MEDLINE | ID: mdl-18237399

ABSTRACT

BACKGROUND: Wound healing in horses is fraught with complications. Specifically, wounds on horse limbs often develop exuberant granulation tissue which behaves clinically like a benign tumor and resembles the human keloid in that the evolving scar is trapped in the proliferative phase of repair, leading to fibrosis. Clues gained from the study of over-scarring in horses should eventually lead to new insights into how to prevent unwanted scar formation in humans. cDNA fragments corresponding to CTNNB1 (coding for beta-catenin) and PECAM1, genes potentially contributing to the proliferative phase of repair, were previously identified in a mRNA expression study as being up-regulated in 7 day wound biopsies from horses. The aim of the present study was to clone full-length equine CTNNB1 and PECAM1 cDNAs and to study the spatio-temporal expression of mRNAs and corresponding proteins during repair of body and limb wounds in a horse model. RESULTS: The temporal pattern of the two genes was similar; except for CTNNB1 in limb wounds, wounding caused up-regulation of mRNA which did not return to baseline by the end of the study. Relative over-expression of both CTNNB1 and PECAM1 mRNA was noted in body wounds compared to limb wounds. Immunostaining for both beta-catenin and PECAM1 was principally observed in endothelial cells and fibroblasts and was especially pronounced in wounds having developed exuberant granulation tissue. CONCLUSION: This study is the first to characterize equine cDNA for CTNNB1 and PECAM1 and to document that these genes are expressed during wound repair in horses. It appears that beta-catenin may be regulated in a post-transcriptional manner while PECAM1 might help thoracic wounds mount an efficient inflammatory response in contrast to what is observed in limb wounds. Furthermore, data from this study suggest that beta-catenin and PECAM1 might interact to modulate endothelial cell and fibroblast proliferation during wound repair in the horse.


Subject(s)
Cicatrix/genetics , Horses/genetics , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Wound Healing/genetics , beta Catenin/genetics , Animals , Base Sequence , Gene Expression/genetics , Gene Expression Regulation/genetics , Molecular Sequence Data , Reference Values
17.
Cancer Lett ; 262(2): 265-75, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18234419

ABSTRACT

The protein tyrosine phosphatase (PTP) superfamily of enzymes functions with protein tyrosine kinases to regulate a broad spectrum of fundamental physiological processes. Addition of the PTP inhibitor potassium bisperoxo(1,10-phenanthroline)oxo-vanadate(V) [bpV(phen)] to the culture medium of human ovarian cancer cells (OVCAR-3) resulted in a dose-dependent decrease in the formation of tumors in a 3-D culture system. An evaluation of the potency of bpV(phen) in vivo confirmed the anti-tumor activity. Further study of the mechanism of action revealed a 40% decrease in Cdk2 kinase activity, an elevated level of Cdk2/p27(kip1), and the appearance of Cdk2/SHP-1 complexes. Therefore, a cytostatic dose of a PTP inhibitor increases the intracellular levels of Cdk2/p27(kip) and Cdk2/SHP-1 complexes, which indicate the presence of additional mechanisms underlying the anti-tumor activity.


Subject(s)
Adenocarcinoma/metabolism , Calcium-Binding Proteins/metabolism , Cyclin-Dependent Kinase 2/metabolism , Ovarian Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein Tyrosine Phosphatases/antagonists & inhibitors , Cell Culture Techniques , Female , Humans , Organometallic Compounds/pharmacology , Phenanthrolines/pharmacology , Tumor Cells, Cultured
18.
Mol Reprod Dev ; 74(12): 1491-504, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17410545

ABSTRACT

In mammals, the pre-Sertoli cell of the male genital ridge is the first cell type to display sex specific differentiation and differential gene expression. The genetic cascade driving the differentiation of pre-Sertoli cells and ultimately testis formation is beginning to be unravelled, but many questions remain. A better understanding of the transcriptome of pre-Sertoli cells immediately after sex determination is essential in order to further understand this differentiation process. A mouse model expressing Red Fluorescent Protein (RFP) under the control of a hybrid mouse/pig SRY promoter (HybSRYp-RFP) was used to purify cells from embryonic day 12.0 (e12.0) male genital ridges. To compare the transcriptomes of HybSRYp-RFP cell populations versus age matched whole female genital ridges, RNA was extracted and used to generate molecular probes that were hybridized onto Affymetrix Mouse Genome 430 2.0 micro-arrays. The expression of genes considered markers for pre-Sertoli cells, including Sox9, Mis, Dhh and Fgf9 were identified within the HybSRYp-RFP expressing cell population, while markers for germ cells (Oct4, SSEA-1) and endothelial cells (Ntrk3) were not identified. In contrast, markers for ovarian somatic cell expression, including Fst and Bmp2, were identified as overexpressed within the ovarian cell population. In a general fashion, genes identified as 2.5-fold over expressed in HybSRYp-RFP expressing cells coded notably for cell signalling and extra cellular proteins. The expression of Sox10, Stc2, Fgf18, Fgf13 and Wnt6 were further characterized via whole mount in situ hybridization (WISH) on male and female genital ridges between e11.5 and e14.5. Sox10, Fgf18, Fgf13 and Stc2 gene expression was detected within the male genital ridges while Wnt6 was found diffusely within both the male and female genital ridges. These data represent the earliest comprehensive microarray expression analysis of purified presumptive pre-Sertoli cells available to date.


Subject(s)
Gene Expression , Organogenesis/genetics , Sertoli Cells/metabolism , Testis/embryology , Animals , Cell Differentiation/genetics , Gene Expression Profiling , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Male , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Signal Transduction/genetics , Testis/metabolism , Red Fluorescent Protein
19.
J Mol Endocrinol ; 38(1-2): 67-78, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17242170

ABSTRACT

The type 1 form of 17beta-hydroxysteroid dehydrogenase (17betaHSD1) was the first isoform to be identified and is capable of converting estrone to 17beta-estradiol. This study was aimed at characterizing the molecular structure of the equine 17betaHSD1 gene and cDNA, as well as its molecular regulation during human chorionic gonadotropin (hCG)-induced follicular luteinization/ovulation in vivo. The equine 17betaHSD1 gene was cloned from an equine genomic library and shown to have a conserved genomic structure composed of six exons. Its cDNA sequence was also identified and coded for a 308 amino acid protein, 72 x 1-74 x 5% homologous to other mammalian orthologs. RT-PCR/Southern blot analyses were performed to study the regulation of the 17betaHSD1 transcript in equine preovulatory follicles isolated between 0 and 39 h after hCG treatment. Results demonstrated the presence of high 17betaHSD1 mRNA expression prior to hCG treatment with a marked decrease observed 12 h after hCG (P<0 x 05). Analyses on isolated preparations of granulosa and theca interna cells identified the granulosa cell layer as the site of 17betaHSD1 transcript expression and downregulation (P<0 x 05). A 1412 bp fragment of the equine 17betaHSD1 proximal promoter was sequenced and shown to contain many putative transcription factor binding sites. Electromobility shift assays (EMSA) using a fragment of the proximal promoter (-230/-30) and nuclear extracts prepared from granulosa cells isolated prior to hCG (0 h post-hCG) revealed the presence of a major complex, and results from competition assays suggest that steroidogenic factor-1 (SF-1), NFkappaB, GATA, and Sp1 cis-elements are involved. Supershift assays using an antibody against the p65 subunit of NFkappaB led to the displacement of the binding nuclear proteins to the DNA probe, whereas the use of an anti-equine SF-1 antibody demonstrated the clear formation of a DNA-protein-antibody complex, confirming the potential role of these transcription factors in EMSA results. Interestingly, a notable decrease in DNA binding was observed when granulosa cell nuclear extracts isolated 30 h post-hCG were used, which paralleled the decrease in 17betaHSD1 transcript after hCG treatment. Thus, this study is the first to report the gonadotropin-dependent downregulation of 17betaHSD1 transcript expression in a monoovulatory species, the presence and regulation of protein/DNA interactions in the proximal region of the 17betaHSD1 promoter during gonadotropin treatment, and the characterization of the primary structure of equine 17betaHSD1 cDNA and gene.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Down-Regulation/physiology , Horses/genetics , Luteinization/physiology , Ovarian Follicle/enzymology , 17-Hydroxysteroid Dehydrogenases/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , DNA-Binding Proteins/physiology , Female , Molecular Sequence Data , Promoter Regions, Genetic
20.
Gene ; 391(1-2): 63-75, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17250974

ABSTRACT

Vacuolar system-associated protein-60 (VASAP-60) constitutes the bovine ortholog of the human "protein kinase C substrate 80K-H" (PRKCSH or 80K-H). We characterized the bovine VASAP-60/PRKCSH gene structure and promoter, identified cis-acting elements controlling VASAP-60 expression, searched for mRNA splice variants, and analyzed mRNA expression in ovarian follicles. Expression of VASAP-60 mRNA showed a 2.4-fold increase (P<0.0001) in granulosa cells of dominant follicles compared to small follicles (2-4 mm) or ovulatory follicles, and no mRNA splice variant was identified. The bovine VASAP-60 gene encompasses 12.5 kb and is composed of 18 exons and 17 introns. Primer extension analysis revealed a single transcription initiation site, and the promoter lacks a TATA box. Promoter activity assays were performed with a series of deletion constructs in different bovine cell lines (endometrial epithelial glandular, kidney epithelial and aortic endothelial) to identify cis-acting elements. The -53/+16 bp fragment (+1 = transcription start site) conferred minimal promoter activity whereas activator and repressor elements were located in the -200/-53 bp and -653/-200 bp fragments, respectively. Analysis of cis-acting elements in the -200/-53 bp activation domain revealed by gel shift assays and chromatin immunoprecipitation assay that transcription factor YY1 binds to VASAP-60 promoter. This study is the first to report that VASAP-60 is up-regulated in granulosa cells of dominant follicles, to document the primary structure of the bovine VASAP-60 gene and promoter, and to demonstrate that YY1 binds to the VASAP-60 proximal promoter and may act as a positive transcriptional regulator.


Subject(s)
Gene Expression Profiling , Membrane Proteins/genetics , Promoter Regions, Genetic/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cell Line , Electrophoretic Mobility Shift Assay , Exons , Female , Granulosa Cells/metabolism , Introns , Luciferases/genetics , Luciferases/metabolism , Male , Molecular Sequence Data , Ovarian Follicle/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Transcription Initiation Site , YY1 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...