Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 10688, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37393374

ABSTRACT

The establishment of left-right patterning in mice occurs at a transient structure called the embryonic node or left-right organizer (LRO). Previous analysis of the LRO has proven challenging due to the small cell number and transient nature of this structure. Here, we seek to overcome these difficulties to define the transcriptome of the LRO. Specifically, we used single cell RNA sequencing of 0-1 somite embryos to identify LRO enriched genes which were compared to bulk RNA sequencing of LRO cells isolated by fluorescent activated cell sorting. Gene ontology analysis indicated an enrichment of genes associated with cilia and laterality terms. Furthermore, comparison to previously identified LRO genes identified 127 novel LRO genes, including Ttll3, Syne1 and Sparcl1, for which the expression patterns were validated using whole mount in situ hybridization. This list of novel LRO genes will be a useful resource for further studies on LRO morphogenesis, the establishment of laterality and the genetic causes of heterotaxy.


Subject(s)
Cilia , Transcriptome , Animals , Mice , Cell Count , Cell Separation , Cilia/genetics , RNA , Calcium-Binding Proteins , Extracellular Matrix Proteins
2.
Hum Mol Genet ; 32(14): 2335-2346, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37158461

ABSTRACT

FOXJ1 is expressed in ciliated cells of the airways, testis, oviduct, central nervous system and the embryonic left-right organizer. Ablation or targeted mutation of Foxj1 in mice, zebrafish and frogs results in loss of ciliary motility and/or reduced length and number of motile cilia, affecting the establishment of the left-right axis. In humans, heterozygous pathogenic variants in FOXJ1 cause ciliopathy leading to situs inversus, obstructive hydrocephalus and chronic airway disease. Here, we report a novel truncating FOXJ1 variant (c.784_799dup; p.Glu267Glyfs*12) identified by clinical exome sequencing from a patient with isolated congenital heart defects (CHD) which included atrial and ventricular septal defects, double outlet right ventricle (DORV) and transposition of the great arteries. Functional experiments show that FOXJ1 c.784_799dup; p.Glu267Glyfs*12, unlike FOXJ1, fails to induce ectopic cilia in frog epidermis in vivo or to activate the ADGB promoter, a downstream target of FOXJ1 in cilia, in transactivation assays in vitro. Variant analysis of patients with heterotaxy or heterotaxy-related CHD indicates that pathogenic variants in FOXJ1 are an infrequent cause of heterotaxy. Finally, we characterize embryonic-stage CHD in Foxj1 loss-of-function mice, demonstrating randomized heart looping. Abnormal heart looping includes reversed looping (dextrocardia), ventral looping and no looping/single ventricle hearts. Complex CHDs revealed by histological analysis include atrioventricular septal defects, DORV, single ventricle defects as well as abnormal position of the great arteries. These results indicate that pathogenic variants in FOXJ1 can cause isolated CHD.


Subject(s)
Heart Defects, Congenital , Heart Septal Defects , Heterotaxy Syndrome , Transposition of Great Vessels , Humans , Male , Forkhead Transcription Factors/genetics , Heart Atria , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Transposition of Great Vessels/genetics
3.
Curr Opin Genet Dev ; 75: 101937, 2022 08.
Article in English | MEDLINE | ID: mdl-35777348

ABSTRACT

Heterotaxy is a disorder with complex congenital heart defects and diverse left-right (LR) patterning defects in other organ systems. Despite evidence suggesting a strong genetic component in heterotaxy, the majority of molecular causes remain unknown. Established genes often involve a ciliated, embryonic structure known as the left-right organizer (LRO). Herein, we focus on genetic discoveries in heterotaxy in the past two years. These include complex genetic architecture, novel mechanisms regulating cilia formation, and evidence for conservation of LR patterning between distant species. We feature new insights regarding established LR signaling pathways, bring attention to heterotaxy candidate genes in novel pathways, and provide an extensive overview of genes previously associated with laterality phenotypes in humans.


Subject(s)
Body Patterning , Heterotaxy Syndrome , Body Patterning/genetics , Cilia/metabolism , Heterotaxy Syndrome/genetics , Humans , Phenotype , Signal Transduction/genetics
4.
Breast Cancer Res ; 20(1): 35, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720215

ABSTRACT

BACKGROUND: The majority of estrogen receptor-positive (ERα+) breast cancers respond to endocrine therapies. However, resistance to endocrine therapies is common in 30% of cases, which may be due to altered ERα signaling and/or enhanced plasticity of cancer cells leading to breast cancer subtype conversion. The mechanisms leading to enhanced plasticity of ERα-positive cancer cells are unknown. METHODS: We used short hairpin (sh)RNA and/or the CRISPR/Cas9 system to knockdown the expression of the dependence receptor UNC5A in ERα+ MCF7 and T-47D cell lines. RNA-seq, quantitative reverse transcription polymerase chain reaction, chromatin immunoprecipitation, and Western blotting were used to measure the effect of UNC5A knockdown on basal and estradiol (E2)-regulated gene expression. Mammosphere assay, flow cytometry, and immunofluorescence were used to determine the role of UNC5A in restricting plasticity. Xenograft models were used to measure the effect of UNC5A knockdown on tumor growth and metastasis. Tissue microarray and immunohistochemistry were utilized to determine the prognostic value of UNC5A in breast cancer. Log-rank test, one-way, and two-way analysis of variance (ANOVA) were used for statistical analyses. RESULTS: Knockdown of the E2-inducible UNC5A resulted in altered basal gene expression affecting plasma membrane integrity and ERα signaling, as evident from ligand-independent activity of ERα, altered turnover of phosphorylated ERα, unique E2-dependent expression of genes effecting histone demethylase activity, enhanced upregulation of E2-inducible genes such as BCL2, and E2-independent tumorigenesis accompanied by multiorgan metastases. UNC5A depletion led to the appearance of a luminal/basal hybrid phenotype supported by elevated expression of basal/stem cell-enriched ∆Np63, CD44, CD49f, epidermal growth factor receptor (EGFR), and the lymphatic vessel permeability factor NTN4, but lower expression of luminal/alveolar differentiation-associated ELF5 while maintaining functional ERα. In addition, UNC5A-depleted cells acquired bipotent luminal progenitor characteristics based on KRT14+/KRT19+ and CD49f+/EpCAM+ phenotype. Consistent with in vitro results, UNC5A expression negatively correlated with EGFR expression in breast tumors, and lower expression of UNC5A, particularly in ERα+/PR+/HER2- tumors, was associated with poor outcome. CONCLUSION: These studies reveal an unexpected role of the axon guidance receptor UNC5A in fine-tuning ERα and EGFR signaling and the luminal progenitor status of hormone-sensitive breast cancers. Furthermore, UNC5A knockdown cells provide an ideal model system to investigate metastasis of ERα+ breast cancers.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Receptors, Cell Surface/genetics , Breast Neoplasms/pathology , CRISPR-Cas Systems/genetics , Carcinogenesis/genetics , Cell Plasticity/genetics , ErbB Receptors/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , MCF-7 Cells , Neoplasm Metastasis , Netrin Receptors , RNA, Small Interfering/genetics
5.
Mol Cancer Ther ; 16(12): 2747-2758, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28978719

ABSTRACT

Breast cancer progression is associated with systemic effects, including functional limitations and sarcopenia without the appearance of overt cachexia. Autocrine/paracrine actions of cytokines/chemokines produced by cancer cells mediate cancer progression and functional limitations. The cytokine-inducible transcription factor NF-κB could be central to this process, as it displays oncogenic functions and is integral to the Pax7:MyoD:Pgc-1ß:miR-486 myogenesis axis. We tested this possibility using the MMTV-PyMT transgenic mammary tumor model and the NF-κB inhibitor dimethylaminoparthenolide (DMAPT). We observed deteriorating physical and functional conditions in PyMT+ mice with disease progression. Compared with wild-type mice, tumor-bearing PyMT+ mice showed decreased fat mass, impaired rotarod performance, and reduced grip strength as well as increased extracellular matrix (ECM) deposition in muscle. Contrary to acute cachexia models described in the literature, mammary tumor progression was associated with reduction in skeletal muscle stem/satellite-specific transcription factor Pax7. Additionally, we observed tumor-induced reduction in Pgc-1ß in muscle, which controls mitochondrial biogenesis. DMAPT treatment starting at 6 to 8 weeks age prior to mammary tumor occurrence delayed mammary tumor onset and tumor growth rates without affecting metastasis. DMAPT overcame cancer-induced functional limitations and improved survival, which was accompanied with restoration of Pax7, Pgc-1ß, and mitochondria levels and reduced ECM levels in skeletal muscles. In addition, DMAPT restored circulating levels of 6 out of 13 cancer-associated cytokines/chemokines changes to levels seen in healthy animals. These results reveal a pharmacological approach for overcoming cancer-induced functional limitations, and the above-noted cancer/drug-induced changes in muscle gene expression could be utilized as biomarkers of functional limitations. Mol Cancer Ther; 16(12); 2747-58. ©2017 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Mammary Neoplasms, Animal/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic
6.
Endocrinology ; 156(7): 2503-17, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25933105

ABSTRACT

The adrenal glands consist of an outer cortex and an inner medulla, and their primary purposes include hormone synthesis and secretion. The adrenal cortex produces a complex array of steroid hormones, whereas the medulla is part of the sympathetic nervous system and produces the catecholamines epinephrine and norepinephrine. In the mouse, GATA binding protein (GATA) 4 and GATA6 transcription factors are coexpressed in several embryonic tissues, including the adrenal cortex. To explore the roles of GATA4 and GATA6 in mouse adrenal development, we conditionally deleted these genes in adrenocortical cells using the Sf1Cre strain of animals. We report here that mice with Sf1Cre-mediated double deletion of Gata4 and Gata6 genes lack identifiable adrenal glands, steroidogenic factor 1-positive cortical cells and steroidogenic gene expression in the adrenal location. The inactivation of the Gata6 gene alone (Sf1Cre;Gata6(flox/flox)) drastically reduced the adrenal size and corticosterone production in the adult animals. Adrenocortical aplasia is expected to result in the demise of the animal within 2 weeks after birth unless glucocorticoids are provided. In accordance, Sf1Cre;Gata4(flox/flox)Gata6(flox/flox) females depend on steroid supplementation to survive after weaning. Surprisingly, Sf1Cre;Gata4(flox/flox)Gata6(flox/flox) males appear to live normal lifespans as vital steroidogenic synthesis shifts to their testes. Our results reveal a requirement for GATA factors in adrenal development and provide a novel tool to characterize the transcriptional network controlling adrenocortical cell fates.


Subject(s)
Adrenal Glands/embryology , Adrenal Insufficiency/genetics , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , RNA, Messenger/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Adrenal Cortex/embryology , Adrenal Cortex/metabolism , Adrenal Glands/metabolism , Animals , Corticosterone/metabolism , Female , Gene Deletion , Gene Expression , Hypoadrenocorticism, Familial , Male , Mice , Sex Factors , Steroidogenic Factor 1/metabolism , Testis/metabolism
7.
Endocrinology ; 156(5): 1873-86, 2015 May.
Article in English | MEDLINE | ID: mdl-25668066

ABSTRACT

The roles of the GATA4 and GATA6 transcription factors in testis development were examined by simultaneously ablating Gata4 and Gata6 with Sf1Cre (Nr5a1Cre). The deletion of both genes resulted in a striking testicular phenotype. Embryonic Sf1Cre; Gata4(flox/flox) Gata6(flox/flox) (conditional double mutant) testes were smaller than control organs and contained irregular testis cords and fewer gonocytes. Gene expression analysis revealed significant down-regulation of Dmrt1 and Mvh. Surprisingly, Amh expression was strongly up-regulated and remained high beyond postnatal day 7, when it is normally extinguished. Neither DMRT1 nor GATA1 was detected in the Sertoli cells of the mutant postnatal testes. Furthermore, the expression of the steroidogenic genes Star, Cyp11a1, Hsd3b1, and Hsd17b3 was low throughout embryogenesis. Immunohistochemical analysis revealed a prominent reduction in cytochrome P450 side-chain cleavage enzyme (CYP11A1)- and 3ß-hydroxysteroid dehydrogenase-positive (3ßHSD) cells, with few 17α-hydroxylase/17,20 lyase-positive (CYP17A1) cells present. In contrast, in postnatal Sf1Cre; Gata4(flox/flox) Gata6(flox/flox) testes, the expression of the steroidogenic markers Star, Cyp11a1, and Hsd3b6 was increased, but a dramatic down-regulation of Hsd17b3, which is required for testosterone synthesis, was observed. The genes encoding adrenal enzymes Cyp21a1, Cyp11b1, Cyp11b2, and Mcr2 were strongly up-regulated, and clusters containing numerous CYP21A2-positive cells were localized in the interstitium. These data suggest a lack of testis functionality, with a loss of normal steroidogenic testis function, concomitant with an expansion of the adrenal-like cell population in postnatal conditional double mutant testes. Sf1Cre; Gata4(flox/flox) Gata6(flox/flox) animals of both sexes lack adrenal glands; however, despite this deficiency, males are viable in contrast to the females of the same genotype, which die shortly after birth.


Subject(s)
GATA4 Transcription Factor/physiology , GATA6 Transcription Factor/physiology , RNA, Messenger/metabolism , Testis/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Adrenal Glands/metabolism , Animals , Anti-Mullerian Hormone/genetics , Anti-Mullerian Hormone/metabolism , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Developmental , Male , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Steroid 21-Hydroxylase/genetics , Steroid 21-Hydroxylase/metabolism , Testis/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Biol Reprod ; 91(1): 24, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24899573

ABSTRACT

Granulosa cell formation and subsequent follicular assembly are important for ovarian development and function. Two members of the GATA family of transcription factors, GATA4 and GATA6, are expressed in ovarian somatic cells early in development, and their importance in adult ovarian function has been recently highlighted. In this study, we demonstrated that the embryonic loss of Gata4 and Gata6 expression within the ovary results in a strong down-regulation of genes involved in the ovarian developmental pathway (Fst and Irx3) as well as diminished expression of the pregranulosa and granulosa cell markers SPRR2 and FOXL2, respectively. Postnatal ovaries deficient in both Gata genes show impaired somatic cell proliferation and arrested follicular development at the primordial stage, where oocytes are either enclosed by one layer of squamous granulosa cells or remain in germ cell nests/clusters. Furthermore, germ cell nests and primordial follicles are predominantly localized to the central region of the Sf1Cre; Gata4(flox/flox) Gata6(flox/flox) ovaries, where the boundary between the medulla and cortex is almost nonexistent. Lastly, most of the oocytes are lost early in development in conditional double mutant ovaries, which confirms the importance of normally differentiated granulosa cells as supporting cells for oocyte survival. Thus, both GATA4 and GATA6 proteins are fundamental regulators of granulosa cell differentiation and proliferation, and consequently of proper follicular assembly during normal ovarian development and function.


Subject(s)
GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , Oocytes/metabolism , Ovarian Follicle/growth & development , Ovary/metabolism , Animals , Down-Regulation , Female , GATA4 Transcription Factor/metabolism , GATA6 Transcription Factor/metabolism , Gene Deletion , Granulosa Cells/cytology , Granulosa Cells/metabolism , Mice , Oocytes/cytology , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovary/cytology
9.
Dev Biol ; 381(1): 144-58, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23769843

ABSTRACT

Sex determination in mammals requires interaction between the transcription factor GATA4 and its cofactor FOG2. We have recently described the function of both proteins in testis development beyond the sex determination stage; their roles in the postnatal ovary, however, remain to be defined. Here, we use gene targeting in mice to determine the requirement of GATA4 and FOG2 in ovarian development and folliculogenesis. The results from this study identify an essential role of the GATA4 protein in the ovarian morphogenetic program. We show that in contrast to the sex determination phase, which relies on the GATA4-FOG2 complex, the subsequent regulation of ovarian differentiation is dependent upon GATA4 but not FOG2. The loss of Gata4 expression within the ovary results in impaired granulosa cell proliferation and theca cell recruitment as well as fewer primordial follicles in the ovarian cortex, causing a failure in follicular development. Preantral follicular atresia is observed within the few follicles that develop despite Gata4 deficiency. The depletion of the follicular pool in GATA4 deficient ovary results in the formation of ovarian cysts and sterility.


Subject(s)
DNA-Binding Proteins/metabolism , GATA4 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Ovarian Follicle/embryology , Ovary/embryology , Transcription Factors/metabolism , Alleles , Animals , Cell Differentiation , Cell Proliferation , Female , Gene Deletion , Hypothalamus/metabolism , Mice , Mice, Inbred C57BL , Mutation , Ovarian Cysts/metabolism , Ovary/metabolism , Pituitary Gland/metabolism , Zinc Fingers
10.
Biol Reprod ; 86(4): 123, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22278982

ABSTRACT

Type 5 acid phosphatase (ACP5; also known as tartrate-resistant acid phosphatase or uteroferrin) is a metalloprotein secreted by the endometrial glandular epithelium of pigs, mares, sheep, and water buffalo. In this paper, we describe the phylogenetic distribution of endometrial expression of ACP5 and demonstrate that endometrial expression arose early in evolution (i.e., before divergence of prototherian and therian mammals ~166 million years ago). To determine expression of ACP5 in the pregnant endometrium, RNA was isolated from rhesus, mouse, rat, dog, sheep, cow, horse, armadillo, opossum, and duck-billed platypus. Results from RT-PCR and RNA-Seq experiments confirmed that ACP5 is expressed in all species examined. ACP5 was also demonstrated immunochemically in endometrium of rhesus, marmoset, sheep, cow, goat, and opossum. Alignment of inferred amino acid sequences shows a high conservation of ACP5 throughout speciation, with species-specific differences most extensive in the N-terminal and C-terminal regions of the protein. Analysis by Selecton indicated that most of the sites in ACP5 are undergoing purifying selection, and no sites undergoing positive selection were found. In conclusion, endometrial expression of ACP5 is a common feature in all orders of mammals and has been subjected to purifying selection. Expression of ACP5 in the uterus predates the divergence of therians and prototherians. ACP5 is an evolutionary conserved gene that likely exerts a common function important for pregnancy in mammals using a wide range of reproductive strategies.


Subject(s)
Acid Phosphatase/metabolism , Biological Evolution , Endometrium/metabolism , Isoenzymes/metabolism , Mammals/genetics , Pregnancy, Animal/genetics , Acid Phosphatase/genetics , Amino Acid Sequence , Animals , Female , Isoenzymes/genetics , Mammals/physiology , Molecular Sequence Data , Phylogeny , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Tartrate-Resistant Acid Phosphatase
11.
Am J Reprod Immunol ; 64(4): 265-74, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20678169

ABSTRACT

Uterine serpins (recently designated as SERPINA14) are hormonally induced proteins secreted in large quantities by the endometrial epithelium during pregnancy. The SERPINA14 proteins belong to the serine proteinase inhibitor (serpin) superfamily, but their apparent lack of inhibitory activity toward serine proteinases suggests that these proteins evolved a different function from the anti-proteinase activity typically found in most members of the serpin superfamily. The gene is present in a limited group of mammals in the Laurasiatheria superorder (ruminants, horses, pigs, dolphins and some carnivores) while being absent in primates, rodents, lagomorphs and marsupials. Thus, the gene is likely to have evolved by gene duplication after divergence of Laurasiatheria and to play an important role in pregnancy. That role may vary between species. In sheep, SERPINA14 probably serves an immunoregulatory role to prevent rejection of the fetal allograft. It is inhibitory to lymphocyte proliferation and natural killer cell function. In the pig, SERPINA14 is involved in iron transport to the fetus by binding to and stabilizing the iron-binding protein uteroferrin. It is possible that SERPINA14 has undergone divergence in function since the original emergence of the gene in a common ancestor of species possessing SERPINA14.


Subject(s)
Endometrium/metabolism , Evolution, Molecular , Fetus/physiology , Mammals/physiology , Pregnancy, Animal , Serpins/genetics , Serpins/physiology , Uterus/metabolism , Animals , Animals, Domestic , Female , Fetus/immunology , Gene Expression Regulation , Mammals/genetics , Mammals/immunology , Mammals/metabolism , Placenta/physiology , Pregnancy , Pregnancy, Animal/immunology , Pregnancy, Animal/metabolism , Pregnancy, Animal/physiology , Progesterone/immunology , Serine Proteinase Inhibitors/metabolism , Serpins/chemistry
12.
FASEB J ; 24(2): 526-37, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19825977

ABSTRACT

Uterine serpins (USs), designated as SERPINA14, are expressed in the endometrium in response to progesterone. All species identified as having USs exhibit epitheliochorial placentation and are in the Ruminantia and Suidae orders of the Laurasiatheria superorder. The objective was to identify US genes in species within and outside Laurasiatheria and evaluate whether evolution of the US gene was associated with development of the epitheliochorial placenta. Through queries of nucleotide and genomic databases, known US genes were identified (caprine, bovine, porcine, water buffalo), and new US coding sequences were found in dolphins, horses, dogs, and cats. The cat sequence contained several stop codons. No sequence was found in completed genomic sequences for primates, rodents, rabbits, opossums, or duck-billed platypuses. Reverse transcription-polymerase chain reaction confirmed expression of the US gene in the uterus of pregnant horses and dogs. The ratio of nonsynonymous/synonymous substitutions suggests that the US gene evolved under positive selection. In conclusion, the US gene evolved within the Laurasiatheria superorder to play a role in pregnancy for species with epitheliochorial placentation and some but not all Laurasiatheria species that have a different form of placentation. The positive selection taking place in the gene suggests development of species-specific functions.


Subject(s)
Evolution, Molecular , Placentation/genetics , Serpins/genetics , Uterus/physiology , Amino Acid Sequence , Animals , Buffaloes/genetics , Cats , Cattle , Dogs , Dolphins/genetics , Female , Goats/genetics , Horses/genetics , Humans , Molecular Sequence Data , Phylogeny , Pregnancy , Sequence Alignment , Serpins/metabolism , Swine/genetics , alpha 1-Antitrypsin/genetics
13.
J Cell Biochem ; 107(6): 1182-8, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19530225

ABSTRACT

The hormonal-regulated serpin, ovine uterine serpin (OvUS), also called uterine milk protein (UTMP), inhibits proliferation of lymphocytes and prostate cancer (PC-3) cells by blocking cell-cycle progression. The present aim was to identify cell-cycle-related genes regulated by OvUS in PC-3 cells using the quantitative human cell-cycle RT(2) Profiler PCR array. Cells were cultured +/-200 microg/ml recombinant OvUS (rOvUS) for 12 and 24 h. At 12 h, rOvUS increased expression of three genes related to cell-cycle checkpoints and arrest (CDKN1A, CDKN2B, and CCNG2). Also, 14 genes were down-regulated including genes involved in progression through S (MCM3, MCM5, PCNA), M (CDC2, CKS2, CCNH, BIRC5, MAD2L1, MAD2L2), G(1) (CDK4, CUL1, CDKN3) and DNA damage checkpoint and repair genes RAD1 and RBPP8. At 24 h, rOvUS decreased expression of 16 genes related to regulation and progression through M (BIRC5, CCNB1, CKS2, CDK5RAP1, CDC20, E2F4, MAD2L2) and G(1) (CDK4, CDKN3, TFDP2), DNA damage checkpoints and repair (RAD17, BRCA1, BCCIP, KPNA2, RAD1). Also, rOvUS down-regulated the cell proliferation marker gene MKI67, which is absent in cells at G(0). Results showed that OvUS blocks cell-cycle progression through upregulation of cell-cycle checkpoint and arrest genes and down-regulation of genes involved in cell-cycle progression.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms/pathology , Serpins/pharmacology , Uterus/chemistry , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Profiling/methods , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Serine Proteinase Inhibitors , Sheep , Up-Regulation
14.
BMC Cell Biol ; 9: 5, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18218135

ABSTRACT

BACKGROUND: Uterine serpins are members of the serine proteinase inhibitor superfamily. Like some other serpins, these proteins do not appear to be functional proteinase inhibitors. The most studied member of the group, ovine uterine serpin (OvUS), inhibits proliferation of several cell types including activated lymphocytes, bovine preimplantation embryos, and cell lines for lymphoma, canine primary osteosarcoma and human prostate cancer (PC-3) cells. The goal for the present study was to evaluate the mechanism by which OvUS inhibits cell proliferation. In particular, it was tested whether inhibition of DNA synthesis in PC-3 cells involves cytotoxic actions of OvUS or the induction of apoptosis. The effect of OvUS in the production of the autocrine and angiogenic cytokine interleukin (IL)-8 by PC-3 cells was also determined. Finally, it was tested whether OvUS blocks specific steps in the cell cycle using both PC-3 cells and lymphocytes. RESULTS: Recombinant OvUS blocked proliferation of PC-3 cells at concentrations as low as 8 mug/ml as determined by measurements of [3H]thymidine incorporation or ATP content per well. Treatment of PC-3 cells with OvUS did not cause cytotoxicity or apoptosis or alter interleukin-8 secretion into medium. Results from flow cytometry experiments showed that OvUS blocked the entry of PC-3 cells into S phase and the exit from G2/M phase. In addition, OvUS blocked entry of lymphocytes into S phase following activation of proliferation with phytohemagglutinin. CONCLUSION: Results indicate that OvUS acts to block cell proliferation through disruption of the cell cycle dynamics rather than induction of cytotoxicity or apoptosis. The finding that OvUS can regulate cell proliferation makes this one of only a few serpins that function to inhibit cell growth.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , DNA/biosynthesis , Prostatic Neoplasms/metabolism , Serpins/pharmacology , Animals , Cell Line, Tumor , Cells, Cultured , DNA Replication , Female , Flow Cytometry , Humans , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , L-Lactate Dehydrogenase/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/pharmacology , Serpins/metabolism , Sheep/metabolism , Uterus/metabolism
15.
Exp Biol Med (Maywood) ; 231(8): 1313-22, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16946400

ABSTRACT

Ovine uterine serpin (OvUS) is produced in the uterus of sheep under the influence of progesterone. It weakly inhibits pepsin and reduces proliferation of lymphocytes, tumor cell lines, and preimplantation embryos. When purified from uterine fluid, the concentration required for its antiproliferative effect in vitro is approximately 0.25-1 mg/ml. Here we show that recombinant (r) OvUS is a more potent regulator of cell proliferation than native (n) OvUS purified from uterine fluid. To produce rOvUS, RNA was extracted from endometrium from a pregnant ewe and cDNA was amplified by reverse transcription-polymerase chain reaction using gene-specific primers. The purified OvUS cDNA was inserted into the ampicillin-resistant plasmid vector pcDNA3.1/ V5-His-TOPO. The plasmid was introduced into the TOP10 Escherichia coli strain, purified, and used for transfection of Freestyle 293-F cells. Digestion of rOvUS with protein N-glycosidase F confirmed that rOvUS was N-glycosylated. Both rOvUS and nOvUS inhibited proliferation of phytohemagglutin-activated sheep lymphocytes and the P388D1 mouse lymphoma and PC-3 prostate cell lines. Inhibition was greater for rOvUS than for nOvUS, and concentrations as low as 15 microg/ml rOvUS were effective at reducing lymphocyte proliferation. Addition of rOvUS to fertilized bovine embryos reduced the cleavage rate and the percentage of embryos that became blastocysts. Native OvUS did not affect cleavage rate and had a smaller effect on development to the blastocyst stage. Experiments demonstrate that OvUS is a more potent inhibitor of cell proliferation and embryonic development than previously believed and add credence to the putative role for the protein in regulating cell proliferation.


Subject(s)
Recombinant Proteins/chemical synthesis , Recombinant Proteins/pharmacology , Serpins/pharmacology , Uterus/metabolism , Animals , Base Sequence , Blotting, Western , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Cloning, Molecular , Embryo, Mammalian/drug effects , Female , Humans , Lymphocytes/drug effects , Mice , Molecular Sequence Data , Pregnancy , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serpins/genetics , Sheep
16.
Mol Reprod Dev ; 71(3): 347-57, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15803459

ABSTRACT

In ewes, the uterine gland knockout (UGKO) phenotype is caused by neonatal exposure to norgestomet to arrest uterine gland development and produce an adult which has a uterus characterized by the lack of endometrial glands. Since endometrial glands in the sheep produce the lymphocyte-inhibitory protein, ovine uterine serpin (OvUS), an experiment was conducted with ewes of the UGKO phenotype to evaluate whether the inhibitory actions of progesterone on tissue rejection responses in utero are dependent upon the presence of endometrial glands. Control and UGKO ewes were ovariectomized and subsequently treated with either 100 mg/day progesterone or corn oil vehicle for 30 days. An autograft and allograft of skin were then placed in each uterine lumen and treatments were continued for an additional 30 days before grafts were examined for survival. All autografts survived and had a healthy appearance after histological analysis. Allografts were generally rejected in ewes treated with vehicle but were present for hormone-treated ewes, regardless of uterine phenotype. Analysis of the histoarchitecture and protein synthetic capacity of the uterus revealed that progesterone induced differentiation of endometrial glands and synthesis and secretion of OvUS in UGKO ewes. The UGKO ewes had reduced density of CD45R+ lymphocytes in the endometrial epithelium and there was a tendency for progesterone to reduce this effect in luminal epithelium. Taken together, results confirm the actions of progesterone to inhibit graft rejection response in utero. Responses of UGKO ewes to progesterone indicate that the hormone can induce de novo development and differentiation of endometrial glands, at least when skin grafts are in the uterus.


Subject(s)
Endometrium/immunology , Exocrine Glands/immunology , Graft Survival/drug effects , Immunosuppression Therapy , Progesterone/administration & dosage , Skin Transplantation , Animals , Female , Sheep, Domestic/genetics , Sheep, Domestic/immunology , Transplantation, Autologous , Transplantation, Homologous
17.
Am J Reprod Immunol ; 53(3): 136-43, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15727568

ABSTRACT

PROBLEM: Ovine uterine serpin (OvUS) is a member of the serine proteinase inhibitor superfamily and is the major protein produced by luminal and glandular epithelium of the sheep endometrium during mid to late pregnancy. The protein does not have prototypical proteinase inhibitory activity but can inhibit a wide variety of lymphocyte functions such as mitogen-induced proliferation and natural killer cell cytotoxicity. METHOD OF STUDY: The antiproliferative actions of OvUS were studied. RESULTS: It was demonstrated that, in addition to inhibiting lymphocyte proliferation, OvUS inhibits growth of two tumor cell lines (D17 and PC-3). The protein also interrupts development of pre-implantation embryos. Inhibition of cell proliferation is not universal, however, as OvUS did not inhibit growth of two non-tumorigenic cell lines (MDBK and BEND). The mechanism of action of inhibitory effects of OvUS is not known although experiments with inhibitors of protein kinase A indicate that the protein does not inhibit lymphocyte proliferation through this pathway. Moreover, the protein does not induce apoptosis. CONCLUSIONS: The finding that OvUS has antiproliferative activity is demonstrative of the wide range of functions exerted by members of the serpin superfamily. The antiproliferative property of OvUS may reflect the role of the protein during pregnancy and may be exploitable for design of new antiproliferative drugs.


Subject(s)
Cell Proliferation/drug effects , Serine Proteinase Inhibitors/pharmacology , Serpins/pharmacology , Sheep/physiology , Uterus/physiology , Animals , Apoptosis/drug effects , Blastocyst/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Female , Humans , Serine Proteinase Inhibitors/isolation & purification , Serpins/isolation & purification
18.
Mol Reprod Dev ; 70(3): 262-70, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15625696

ABSTRACT

The uterine serpins have been described in sheep, cattle, and pigs as a highly diverged group of the large superfamily of serpin proteins that typically function as serine proteinase inhibitors. Here, the range of species that possess and express a uterine serpin gene is extended to the goat. Sequencing of cDNA amplified from total RNA from a pregnant goat at day 25 of pregnancy resulted in a 1,292 bp full-length consensus cDNA sequence for caprine uterine serpin (CaUS). The predicted amino acid sequence of the caprine precursor showed 96%, 82%, 55%, and 56% identity to OvUS, BoUS, PoUS1, and PoUS2, respectively. The signal peptide extends from amino acids 1 to 25, resulting in a secreted protein of 404 amino acids and 46,227 Mr (excluding carbohydrate). Both the goat and sheep uterine serpins have a nine amino acid insert in the Helix I region that is not found in bovine or porcine uterine serpins. A total of 13 amino acids in CaUS are different than those for the nearest homologue, ovine uterine serpin. One of these is in the site of cleavage of the signal sequence, where a single nucleotide substitution (G --> C) changed the cysteine for the sheep, bovine, and porcine genes to a serine. In addition, the amino acid at the putative P1-P1' site (the scissile bond for antiproteinase activity) is a valine for CaUS, BoUS, PoUS1, and PoUS2 versus an alanine for OvUS. The hinge region of all five of the uterine serpins (P17-P9) is distinct from the consensus pattern for inhibitory sequences and it is unlikely, therefore, that the uterine serpins possess prototypical proteinase inhibitory activity. The goat uterine serpin was immunolocalized to the glandular epithelium of the endometrium from a pregnant nanny at day 25 of pregnancy. There was also immunoreactive product in scattered luminal epithelial cells. No immunoreaction product was detected in endometrium from a nanny at day 5 of the estrous cycle. Western blotting of uterine fluid collected from the pregnant uterine horn of a unilaterally-pregnant goat revealed the presence of a protein band at Mr approximately 56,000 that reacted with monoclonal antibody to OvUS. In conclusion, the range of species in which uterine serpins are present and expressed in the uterus includes the goat in addition to the previously described sheep, cow, and pig. In all of these species, the uterine serpin is derived primarily from glandular epithelium, is secreted into the uterine lumen, and contains sequence characteristics suggesting it is not an inhibitory serpin.


Subject(s)
Goats/genetics , Serpins/genetics , Uterus/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cloning, Molecular , DNA Primers , DNA, Complementary/genetics , Endometrium/metabolism , Female , Goats/metabolism , Immunohistochemistry , Molecular Sequence Data , Pregnancy , Sequence Alignment , Sequence Analysis, DNA , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...