Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Proc Natl Acad Sci U S A ; 121(4): e2318093121, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38232291

ABSTRACT

In this study, we aimed to address the current limitations of therapies for macro-metastatic triple-negative breast cancer (TNBC) and provide a therapeutic lead that overcomes the high degree of heterogeneity associated with this disease. Specifically, we focused on well-documented but clinically underexploited cancer-fueling perturbations in mRNA translation as a potential therapeutic vulnerability. We therefore developed an orally bioavailable rocaglate-based molecule, MG-002, which hinders ribosome recruitment and scanning via unscheduled and non-productive RNA clamping by the eukaryotic translation initiation factor (eIF) 4A RNA helicase. We demonstrate that MG-002 potently inhibits mRNA translation and primary TNBC tumor growth without causing overt toxicity in mice. Importantly, given that metastatic spread is a major cause of mortality in TNBC, we show that MG-002 attenuates metastasis in pre-clinical models. We report on MG-002, a rocaglate that shows superior properties relative to existing eIF4A inhibitors in pre-clinical models. Our study also paves the way for future clinical trials exploring the potential of MG-002 in TNBC and other oncological indications.


Subject(s)
RNA Helicases , Triple Negative Breast Neoplasms , Humans , Animals , Mice , RNA Helicases/genetics , RNA Helicases/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Protein Biosynthesis , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Ribosomes/metabolism
2.
Development ; 146(20)2019 10 18.
Article in English | MEDLINE | ID: mdl-31575647

ABSTRACT

WNT signaling plays essential roles in the development and function of the female reproductive tract. Although crosstalk with the Hippo pathway is a key regulator of WNT signaling, whether Hippo itself plays a role in female reproductive biology remains largely unknown. Here, we show that conditional deletion of the key Hippo kinases Lats1 and Lats2 in mouse Müllerian duct mesenchyme cells caused them to adopt the myofibroblast cell fate, resulting in profound reproductive tract developmental defects and sterility. Myofibroblast differentiation was attributed to increased YAP and TAZ expression (but not to altered WNT signaling), leading to the direct transcriptional upregulation of Ctgf and the activation of the myofibroblast genetic program. Müllerian duct mesenchyme cells also became myofibroblasts in male mutant embryos, which impeded the development of the male reproductive tract and resulted in cryptorchidism. The inactivation of Lats1/2 in differentiated uterine stromal cells in vitro did not compromise their ability to decidualize, suggesting that Hippo is dispensable during implantation. We conclude that Hippo signaling is required to suppress the myofibroblast genetic program and maintain multipotency in Müllerian mesenchyme cells.


Subject(s)
Mullerian Ducts/cytology , Mullerian Ducts/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Chromatin Immunoprecipitation , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Endometrium/cytology , Endometrium/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/cytology , Myofibroblasts/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics
3.
Cell Mol Life Sci ; 78(19-20): 6709-6719, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34559254

ABSTRACT

Eukaryotic initiation factor (eIF) 4F plays a central role in the ribosome recruitment phase of cap-dependent translation. This heterotrimeric complex consists of a cap binding subunit (eIF4E), a DEAD-box RNA helicase (eIF4A), and a large bridging protein (eIF4G). In mammalian cells, there are two genes encoding eIF4A (eIF4A1 and eIF4A2) and eIF4G (eIF4G1 and eIF4G3) paralogs that can assemble into eIF4F complexes. To query the essential nature of the eIF4F subunits in normal development, we used CRISPR/Cas9 to generate mouse strains with targeted ablation of each gene encoding the different eIF4F subunits. We find that Eif4e, Eif4g1, and Eif4a1 are essential for viability in the mouse, whereas Eif4g3 and Eif4a2 are not. However, Eif4g3 and Eif4a2 do play essential roles in spermatogenesis. Crossing of these strains to the lymphoma-prone Eµ-Myc mouse model revealed that heterozygosity at the Eif4e or Eif4a1 loci significantly delayed tumor onset. Lastly, tumors derived from Eif4e∆38 fs/+/Eµ-Myc or Eif4a1∆5 fs/+/Eµ-Myc mice show increased sensitivity to the chemotherapeutic agent doxorubicin, in vivo. Our study reveals that eIF4A2 and eIF4G3 play non-essential roles in gene expression regulation during embryogenesis; whereas reductions in eIF4E or eIF4A1 levels are protective against tumor development in a murine Myc-driven lymphoma setting.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Eukaryotic Initiation Factor-4F/genetics , Animals , Female , Gene Expression Regulation/genetics , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Subunits/genetics , Spermatogenesis/genetics
4.
Genes Dev ; 27(23): 2602-14, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24298059

ABSTRACT

RNAi combined with next-generation sequencing has proven to be a powerful and cost-effective genetic screening platform in mammalian cells. Still, this technology has its limitations and is incompatible with in situ mutagenesis screens on a genome-wide scale. Using p53 as a proof-of-principle target, we readapted the CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR associated 9) genome-editing system to demonstrate the feasibility of this methodology for targeted gene disruption positive selection assays. By using novel "all-in-one" lentiviral and retroviral delivery vectors heterologously expressing both a codon-optimized Cas9 and its synthetic guide RNA (sgRNA), we show robust selection for the CRISPR-modified Trp53 locus following drug treatment. Furthermore, by linking Cas9 expression to GFP fluorescence, we use an "all-in-one" system to track disrupted Trp53 in chemoresistant lymphomas in the Eµ-myc mouse model. Deep sequencing analysis of the tumor-derived endogenous Cas9-modified Trp53 locus revealed a wide spectrum of mutants that were enriched with seemingly limited off-target effects. Taken together, these results establish Cas9 genome editing as a powerful and practical approach for positive in situ genetic screens.


Subject(s)
CRISPR-Associated Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Genetic Techniques , Animals , CRISPR-Associated Proteins/metabolism , Female , Gene Targeting , Genes, p53/genetics , Genome/genetics , INDEL Mutation/genetics , Kaplan-Meier Estimate , Lentivirus/genetics , Lymphoma/genetics , Lymphoma/mortality , Lymphoma/therapy , Mice , Mice, Inbred C57BL , Mutation , Reproducibility of Results
5.
Biol Reprod ; 102(5): 1055-1064, 2020 04 24.
Article in English | MEDLINE | ID: mdl-31930396

ABSTRACT

Ovarian cancer (OvCa) remains the most common cause of death from gynecological malignancies. Genetically engineered mouse models have been used to study initiation, origin, progression, and/or mechanisms of OvCa. Based on the clinical features of OvCa, we examined a quadruple combination of pathway perturbations including PTEN, TRP53, RB1, and/or CDH1. To characterize the cancer-promoting events in the ovarian surface epithelium (OSE), Amhr2cre/+ mice were used to ablate floxed alleles of Pten, Trp53, and Cdh1, which were crossed with TgK19GT121 mice to inactivate RB1 in KRT19-expressing cells. Inactivation of PTEN, TRP53, and RB1 with or without CDH1 led to the development of type I low-grade OvCa with enlarged serous papillary carcinomas and some high-grade serous carcinomas (HGSCs) in older mice. Initiation of epithelial hyperplasia and micropapillary carcinoma started earlier at 1 month in the triple mutations of Trp53, Pten, and Rb1 mice as compared to 2 months in quadruple mutations of Trp53, Pten, Rb1, and Cdh1 mice, whereas both genotypes eventually developed enlarged proliferating tumors that invaded into the ovary at 3-4 months. Mice with triple and quadruple mutations developed HGSC and/or metastatic tumors, which disseminated into the peritoneal cavity at 4-6 months. In summary, inactivation of PTEN, TRP53, and RB1 initiates OvCa from the OSE. Additional ablation of CDH1 further increased persistence of tumor dissemination and ascites fluid accumulation enhancing peritoneal metastasis.


Subject(s)
Cadherins/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , PTEN Phosphohydrolase/metabolism , Retinoblastoma Binding Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cadherins/genetics , Cell Transformation, Neoplastic , Epithelium/metabolism , Female , Gene Expression Regulation, Neoplastic , Mice , Mice, Inbred Strains , Mice, Knockout , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/secondary , PTEN Phosphohydrolase/genetics , Retinoblastoma Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics
6.
Reproduction ; 160(2): 307-318, 2020 08.
Article in English | MEDLINE | ID: mdl-32520726

ABSTRACT

The development of the Müllerian ducts into the female reproductive tract requires the coordination of multiple signaling pathways that regulate proliferation, apoptosis and differentiation. The Hippo pathway has been reported to interact with several pathways with established roles in Müllerian duct development; yet, its potential roles in reproductive tract development and function remain mostly uncharacterized. The objective of this study was therefore to characterize the roles of the Hippo transcriptional coactivators YAP and TAZ in the female reproductive tract using transgenic mouse models. This report shows that the concomitant conditional inactivation of Yap and Taz in the mouse Müllerian duct mesenchyme results in postnatal developmental defects of the oviduct. Most notably, discontinuities in the myosalpinx layer lead to the progressive formation of cystic dilations of the isthmus. These defects prevented embryo transport and subsequent implantation in older animals, causing infertility. The loss of YAP/TAZ did not appear to affect other biological processes known to be required for the maintenance of oviductal wall integrity, such as TGF-ß/SMAD and Notch signaling and the biogenesis of miRNA, suggesting that the Hippo pathway acts independently of these processes to direct oviduct development. Taken together, these results suggest redundant and essential roles for YAP and TAZ in the postnatal development of the oviduct and the maintenance of its structural integrity.


Subject(s)
Acyltransferases/physiology , Adaptor Proteins, Signal Transducing/physiology , Embryo, Mammalian/cytology , Embryonic Development , Oocytes/cytology , Oviducts/cytology , Animals , Animals, Newborn , Apoptosis , Cell Differentiation , Cell Proliferation , Embryo, Mammalian/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/physiology , Oviducts/physiology , YAP-Signaling Proteins
7.
FASEB J ; 33(10): 10819-10832, 2019 10.
Article in English | MEDLINE | ID: mdl-31268774

ABSTRACT

Recent reports suggest that the Hippo signaling pathway influences ovarian follicle development; however, its exact roles remain unknown. Here, we examined the ovarian functions of the Hippo kinases large tumor suppressors (LATS)1 and 2, which serve to inactivate the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). Inactivation of Lats1/2 in murine granulosa cells either in vitro or in vivo resulted in a loss of granulosa cell morphology, function, and gene expression. Mutant cells further underwent changes in structure and gene expression suggestive of epithelial-to-mesenchymal transition and transdifferentiation into multiple lineages. In vivo, granulosa cell-specific loss of Lats1/2 caused the ovarian parenchyma to be mostly replaced by bone tissue and seminiferous tubule-like structures. Transdifferentiation into Sertoli-like cells and osteoblasts was attributed in part to the increased recruitment of YAP and TAZ to the promoters of sex-determining region Y box 9 and bone γ-carboxyglutamate protein, key mediators of male sex determination and osteogenesis, respectively. Together, these results demonstrate for the first time a critical role for Lats1/2 in the maintenance of the granulosa cell genetic program and further highlight the remarkable plasticity of granulosa cells.-Tsoi, M., Morin, M., Rico, C., Johnson, R. L., Paquet, M., Gévry, N., Boerboom, D. Lats1 and Lats2 are required for ovarian granulosa cell fate maintenance.


Subject(s)
Granulosa Cells/cytology , Granulosa Cells/metabolism , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Lineage , Cell Transdifferentiation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation , Hippo Signaling Pathway , Infertility, Female/genetics , Infertility, Female/pathology , Infertility, Female/physiopathology , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoblasts/pathology , Ovarian Follicle/physiology , Ovary/pathology , Ovary/physiopathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sertoli Cells/metabolism , Sertoli Cells/pathology , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
8.
Biol Reprod ; 100(1): 49-60, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30010727

ABSTRACT

Wnt4 and Wnt5a have well-established roles in the embryonic development of the female reproductive tract, as well as in implantation, decidualization, and ovarian function in adult mice. Although these roles appear to overlap, whether Wnt5a and Wnt4 are functionally redundant in these tissues has not been determined. We addressed this by concomitantly inactivating Wnt4 and Wnt5a in the Müllerian mesenchyme and in ovarian granulosa cells by crossing mice bearing floxed alleles to the Amhr2cre strain. Whereas fertility was reduced by ∼50% in Wnt4flox/flox; Amhr2cre/+ and Wnt5aflox/flox; Amhr2cre/+ females, Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice were either nearly or completely sterile. Loss of fertility was not due to an ovarian defect, as serum ovarian hormone levels, follicle counts, and ovulation rates were comparable to controls. Conversely, the uterus was abnormal in Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice, with thin myometrial and stromal layers, frequent fibrosis and a >90% reduction in numbers of uterine glands, suggesting redundant or additive roles of Wnt4 and Wnt5a in uterine adenogenesis. Loss of fertility in Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice was attributed to defects in decidualization, implantation, and placental development, the severity of which were proportional to the extent of gland loss. Furthermore, a third of Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ females had a partial agenesis of Müllerian duct-derived structures, but with normal oviducts and ovaries. Together, our results suggest that Wnt4 and Wnt5a play redundant roles in the development of the female reproductive tract, and may provide insight into the etiology of certain cases of Müllerian agenesis in women.


Subject(s)
Mullerian Ducts/abnormalities , Mullerian Ducts/metabolism , Urogenital Abnormalities/genetics , Uterus/abnormalities , Wnt-5a Protein/genetics , Wnt4 Protein/genetics , Animals , Female , Gene Deletion , Infertility, Female/embryology , Infertility, Female/genetics , Infertility, Female/metabolism , Male , Mesoderm/abnormalities , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Transgenic , Mullerian Ducts/pathology , Myometrium/abnormalities , Myometrium/metabolism , Pregnancy , Urogenital Abnormalities/metabolism , Urogenital Abnormalities/pathology , Uterus/metabolism , Wnt-5a Protein/metabolism , Wnt4 Protein/metabolism
9.
J Mammary Gland Biol Neoplasia ; 22(3): 203-214, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28822004

ABSTRACT

Canine mammary tumors (CMTs) are the most common neoplasms in intact female dogs. Some clinical and molecular similarities between certain CMT subtypes and breast cancer make them a potential model for the study of the human disease. As misregulated Hippo signaling is thought to play an important role in breast cancer development and also occurs in CMTs, we sought to determine if Hippo represents a valid pharmacological target for the treatment of CMTs. Six CMT cell lines were assessed for their expression of the Hippo pathway effectors YAP and TAZ and for their sensitivity to verteporfin, an inhibitor of YAP-mediated transcriptional coactivation. Four cell lines that expressed YAP (CMT-9, -12, -28, -47) were found to be very sensitive to verteporfin treatment, which killed the cells through induction of apoptosis with ED50 values of 14-79 nM. Conversely, two YAP-negative cell lines (CF-35, CMT-25) were an order of magnitude more resistant to verteporfin. Verteporfin suppressed the expression of YAP/TAZ target genes, particularly CYR61 and CTGF, which play important roles in breast cancer development. Verteporfin was also able to inhibit cell migration and anchorage-independent growth. Likewise, verteporfin efficiently suppressed tumor cell invasiveness in the CMT-28 and -47 lines, but not in CF-35 cells. Together, our findings provide proof of principle that pharmacological targeting of the Hippo pathway compromises the viability and attenuates the malignant behavior of CMT cells. These results will serve as the basis for the development of novel chemotherapeutic approaches for CMTs that could translate to human medicine.


Subject(s)
Mammary Neoplasms, Animal/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Dogs , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Mammary Neoplasms, Animal/drug therapy , Neoplasm Invasiveness/genetics , Porphyrins/pharmacology , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Verteporfin
10.
Biol Reprod ; 97(1): 162-175, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28637242

ABSTRACT

Yes-associated protein (YAP) and WW-containing transcription regulator 1 (WWTR1) are two functionally redundant transcriptional regulators that are downstream effectors of the Hippo signaling pathway, and that act as major regulators of cell growth and differentiation. To elucidate their role in Sertoli cells, primary Sertoli cell culture from Yapflox/flox; Wwtr1flox/flox animals were infected with a Cre recombinase-expressing adenovirus. Concomitant inactivation of Yap and Wwtr1 resulted in a decrease in the mRNA levels of the male sex differentiation genes Dhh, Dmrt1, Sox9, and Wt1, whereas those of genes involved in female differentiation (Wnt4, Rspo1, and Foxl2) were induced. SOX9, FOXL2, and WNT4 proteins were regulated in the same manner as their mRNAs in response to loss of YAP and WWTR1. To further characterize the role of YAP and WWTR1 in Sertoli cells, we generated a mouse model (Yapflox/flox; Wwtr1flox/flox; Amhcre/+) in which Yap and Wwtr1 were conditionally deleted in Sertoli cells. An increase in the number of apoptotic cells was observed in the seminiferous tubules of 4 dpp mutant mice, leading to a reduction in testis weights and a decrease in the number of Sertoli cells in adult animals. Gene expression analyses of testes from 4 dpp Yapflox/flox; Wwtr1flox/flox; Amhcre/+ mice showed that Sertoli cell differentiation is initially altered, as Dhh, Dmrt1, and Sox9 mRNA levels were downregulated, whereas Wnt4 mRNA levels were increased. However, expression of these genes was not changed in older animals. Together, these results suggest a novel role of the Hippo signaling pathway in the mechanisms of sex differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation/physiology , Phosphoproteins/metabolism , Sertoli Cells/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Disorders of Sex Development , Female , Hippo Signaling Pathway , Male , Mice , Mice, Transgenic , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Sperm Count , Trans-Activators , YAP-Signaling Proteins , beta Catenin/genetics , beta Catenin/metabolism
11.
Biol Reprod ; 96(5): 1007-1018, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28339861

ABSTRACT

The luteinizing hormone receptor (LHCGR) is necessary for fertility, and genetic mutations cause defects in reproductive development and function. Activating mutations in LHCGR cause familial male-limited precocious puberty (FMPP). We have previously characterized a mouse model (KiLHRD582G) for FMPP that exhibits the same phenotype of precocious puberty, Leydig cell hyperplasia, and elevated testosterone as boys with the disorder. We observed that KiLHRD582G male mice became infertile by 6 months of age, although sperm count and motility were normal. In this study, we sought to determine the reason for the progressive infertility and the long-term consequences of constant LHCGR signaling. Mating with superovulated females showed that infertile KiLHRD582G mice had functional sperm and normal accessory gland function. Sexual behavior studies revealed that KiLHRD582G mice mounted females, but intromission was brief and ejaculation was not achieved. Histological analysis of the reproductive tract showed unique metaplastic changes resulting in pseudostratified columnar epithelial cells with cilia in the ampulla and chondrocytes in the penile body of the KiLHRD582G mice. The infertile KiLHRD582G exhibited enlarged sinusoids and a decrease in smooth muscle content in the corpora cavernosa of the penile body. However, collagen content was unchanged. Leydig cell adenomas and degenerating seminiferous tubules were seen in 1-year-old KiLHRD582G mice. We conclude that progressive infertility in KiLHRD582G mice is due to sexual dysfunction likely due to functional defects in the penis.


Subject(s)
Adenoma/physiopathology , Infertility, Male/genetics , Infertility, Male/physiopathology , Leydig Cell Tumor/physiopathology , Receptors, LH/genetics , Signal Transduction/genetics , Testicular Neoplasms/physiopathology , Animals , Ejaculation , Estradiol/metabolism , Genitalia, Male/pathology , Genitalia, Male/physiopathology , Infertility, Male/pathology , Leydig Cell Tumor/pathology , Male , Mice , Penis/pathology , Puberty, Precocious/genetics , Sperm Count , Sperm Motility , Testicular Neoplasms/pathology
12.
FASEB J ; 30(4): 1534-47, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26667040

ABSTRACT

Whereas the roles of the canonical wingless-type MMTV (mouse mammary tumor virus) integration site family (WNT) signaling pathway in the regulation of ovarian follicle growth and steroidogenesis are now established, noncanonical WNT signaling in the ovary has been largely overlooked. Noncanonical WNTs, including WNT5a and WNT11, are expressed in granulosa cells (GCs) and are differentially regulated throughout follicle development, but their physiologic roles remain unknown. Using conditional gene targeting, we found that GC-specific inactivation ofWnt5a(but notWnt11) results in the female subfertility associated with increased follicular atresia and decreased rates of ovulation. Microarray analyses have revealed that WNT5a acts to down-regulate the expression of FSH-responsive genesin vitro, and corresponding increases in the expression of these genes have been found in the GCs of conditional knockout mice. Unexpectedly, we found that WNT5a regulates its target genes not by signalingviathe WNT/Ca(2+)or planar cell polarity pathways, but rather by inhibiting the canonical pathway, causing both ß-catenin (CTNNB1) and cAMP responsive element binding (CREB) protein levels to decreaseviaa glycogen synthase kinase-3ß-dependent mechanism. We further found that WNT5a prevents follicle-stimulating hormone and luteinizing protein from up-regulating the CTNNB1 and CREB proteins and their target genes, indicating that WNT5a functions as a physiologic inhibitor of gonadotropin signaling. Together, these findings identify WNT5a as a key regulator of follicle development and gonadotropin responsiveness.-Abedini, A., Zamberlam, G., Lapointe, E., Tourigny, C., Boyer, A., Paquet, M., Hayashi, K., Honda, H., Kikuchi, A., Price, C., Boerboom, D. WNT5a is required for normal ovarian follicle development and antagonizes gonadotropin responsiveness in granulosa cells by suppressing canonical WNT signaling.


Subject(s)
Gonadotropins/pharmacology , Granulosa Cells/drug effects , Ovarian Follicle/metabolism , Wnt Proteins/genetics , Wnt Signaling Pathway/drug effects , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Granulosa Cells/metabolism , Immunoblotting , Immunohistochemistry , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Ovulation/drug effects , Ovulation/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/drug effects , Transcriptome/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , Wnt-5a Protein
13.
PLoS Biol ; 12(3): e1001807, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24618719

ABSTRACT

The Cut homeobox 1 (CUX1) gene is a target of loss-of-heterozygosity in many cancers, yet elevated CUX1 expression is frequently observed and is associated with shorter disease-free survival. The dual role of CUX1 in cancer is illustrated by the fact that most cell lines with CUX1 LOH display amplification of the remaining allele, suggesting that decreased CUX1 expression facilitates tumor development while increased CUX1 expression is needed in tumorigenic cells. Indeed, CUX1 was found in a genome-wide RNAi screen to identify synthetic lethal interactions with oncogenic RAS. Here we show that CUX1 functions in base excision repair as an ancillary factor for the 8-oxoG-DNA glycosylase, OGG1. Single cell gel electrophoresis (comet assay) reveals that Cux1⁺/⁻ MEFs are haploinsufficient for the repair of oxidative DNA damage, whereas elevated CUX1 levels accelerate DNA repair. In vitro base excision repair assays with purified components demonstrate that CUX1 directly stimulates OGG1's enzymatic activity. Elevated reactive oxygen species (ROS) levels in cells with sustained RAS pathway activation can cause cellular senescence. We show that elevated expression of either CUX1 or OGG1 prevents RAS-induced senescence in primary cells, and that CUX1 knockdown is synthetic lethal with oncogenic RAS in human cancer cells. Elevated CUX1 expression in a transgenic mouse model enables the emergence of mammary tumors with spontaneous activating Kras mutations. We confirmed cooperation between Kras(G12V) and CUX1 in a lung tumor model. Cancer cells can overcome the antiproliferative effects of excessive DNA damage by inactivating a DNA damage response pathway such as ATM or p53 signaling. Our findings reveal an alternate mechanism to allow sustained proliferation in RAS-transformed cells through increased DNA base excision repair capability. The heightened dependency of RAS-transformed cells on base excision repair may provide a therapeutic window that could be exploited with drugs that specifically target this pathway.


Subject(s)
DNA Damage , DNA Repair/physiology , Genes, ras/physiology , Homeodomain Proteins/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cellular Senescence/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mammary Neoplasms, Experimental/genetics , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors
14.
Int J Cancer ; 138(11): 2700-12, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26773796

ABSTRACT

In the colon, myofibroblasts are primary contributors in the establishment of the microenvironment involved in tissue homeostasis. Alterations in myofibroblast functions lead to changes resulting in a toxic microenvironment nurturing tumorigenesis. Bone morphogenetic proteins (Bmps) are morphogens known to play key roles in adult gut homeostasis. Studies in genetically-modified mice have shown that Bmp disruption in all cell layers leads to the development of gut polyposis. In contrast, our studies showed that loss of Bmp exclusively in the gastrointestinal epithelium resulted in increased epithelial proliferation without polyposis initiation, thus suggesting a key role for mesenchymal Bmp signaling in polyposis initiation. In order to identify the role of mesenchymal Bmp signaling on the microenvironment and its impact on colonic mucosa, a mouse model was generated with suppression of Bmp signaling exclusively in myofibroblasts (Bmpr1aΔMES). Bmpr1aΔMES mice exhibited increased subepithelial proliferation with changes in cellular composition leading to the development of a primed stroma with modulation of extracellular matrix proteins, immune cells and cytokines as early as 90 days of age. This microenvironmental deregulation was associated with increased polyposis initiation at one year of age. These results are the first to demonstrate that mesenchymal Bmpr1a inactivation alone is sufficient to prompt an expansion of myofibroblasts leading to the development of a reactive mesenchyme that contributes to polyposis initiation in the colon. These findings support the novel concept that inhibition of Bmp signaling in mesenchymal cells surrounding the normal epithelium leads to important changes instructing a toxic microenvironment sufficient to induce colonic polyposis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Colorectal Neoplasms/genetics , Gastrointestinal Neoplasms/genetics , Animals , Animals, Genetically Modified , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Carcinogenesis/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/pathology , Gastrointestinal Neoplasms/pathology , Humans , Mesoderm/growth & development , Mesoderm/pathology , Mice , Mucous Membrane/metabolism , Mucous Membrane/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Microenvironment/genetics
15.
Biol Reprod ; 95(1): 13, 2016 07.
Article in English | MEDLINE | ID: mdl-27281705

ABSTRACT

The mammalian target of rapamycin (Mtor) gene encodes a serine/threonine kinase that acts as a master regulator of processes as diverse as cell growth, protein synthesis, cytoskeleton reorganization, and cell survival. In the testis, physiological roles for Mtor have been proposed in perinatal Sertoli cell proliferation and blood-testis barrier (BTB) remodeling during spermatogenesis, but no in vivo studies of Mtor function have been reported. Here, we used a conditional knockout approach to target Mtor in Sertoli cells. The resulting Mtor(flox/flox); Amhr2(cre/+) mice were characterized by progressive, adult-onset testicular atrophy associated with disorganization of the seminiferous epithelium, loss of Sertoli cell polarity, increased germ cell apoptosis, premature release of germ cells, decreased epididymal sperm counts, increased sperm abnormalities, and infertility. Histopathologic analysis and quantification of the expression of stage-specific markers showed a specific loss of pachytene spermatocytes and spermatids. Although the BTB and the ectoplasmic specializations did not appear to be altered in Mtor(flox/flox);Amhr2(cre/+) mice, a dramatic redistribution of gap junction alpha-1 (GJA1) was detected in their Sertoli cells. Phosphorylation of GJA1 at Ser373, which is associated with its internalization, was increased in the testes of Mtor(flox/flox); Amhr2(cre/+) mice, as was the expression and phosphorylation of AKT, which phosphorylates GJA1 at this site. Together, these results indicate that Mtor expression in Sertoli cells is required for the maintenance of spermatogenesis and the progression of germ cell development through the pachytene spermatocyte stage. One mechanism of mTOR action may be to regulate gap junction dynamics by inhibiting AKT, thereby decreasing GJA1 phosphorylation and internalization. mTOR regulates gap junction alpha-1 protein distribution in Sertoli cells and is necessary for progression through the pachytene spermatocyte stage.


Subject(s)
Connexin 43/metabolism , Gap Junctions/metabolism , Sertoli Cells/metabolism , Spermatogenesis/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Male , Mice , Mice, Knockout , Phosphorylation , Protein Transport/physiology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , Testis/metabolism
16.
Biol Reprod ; 95(4): 76, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27535961

ABSTRACT

Endometriosis causes severe chronic pelvic pain and infertility. Because the standard medication and surgical treatments of endometriosis show high recurrence of symptoms, it is necessary to improve current treatment options. In the initial study, we examined whether niclosamide can be a useful drug for endometriosis in a preclinical setting. Endometriotic implants were induced using an established mouse model involving transimplantation of mouse endometrial fragments to the peritoneal wall of recipient mice. When the recipient mice were treated with niclosamide for 3 weeks, niclosamide reduced the size of endometriotic implants with inhibition of cell proliferation, and inflammatory signaling including RELA (NFKB) and STAT3 activation, but did not alter expression of steroid hormone receptors. To identify genes whose expression is regulated by niclosamide in endometriotic implants, RNA-sequencing was performed, and several genes downregulated by niclosamide were related to inflammatory responses, WNT and MAPK signaling. In a second study designed to assess whether niclosamide affects reproductive function, the recipient mice started receiving niclosamide after the induction of endometriosis. Then, the recipient mice were mated with wild type males, and treatments continued until the pups were born. Niclosamide treated recipient mice became pregnant and produced normal size and number of pups. These results suggest that niclosamide could be an effective therapeutic drug, and acts as an inhibitor of inflammatory signaling without disrupting normal reproductive function.

17.
J Immunol ; 192(1): 259-70, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24285835

ABSTRACT

Salmonella enterica is a ubiquitous Gram-negative intracellular bacterium that continues to pose a global challenge to human health. The etiology of Salmonella pathogenesis is complex and controlled by pathogen, environmental, and host genetic factors. In fact, patients immunodeficient in genes in the IL-12, IL-23/IFN-γ pathway are predisposed to invasive nontyphoidal Salmonella infection. Using a forward genomics approach by N-ethyl-N-nitrosourea (ENU) germline mutagenesis in mice, we identified the Ity14 (Immunity to Typhimurium locus 14) pedigree exhibiting increased susceptibility following in vivo Salmonella challenge. A DNA-binding domain mutation (p.G418_E445) in Stat4 (Signal Transducer and Activator of Transcription Factor 4) was the causative mutation. STAT4 signals downstream of IL-12 to mediate transcriptional regulation of inflammatory immune responses. In mutant Ity14 mice, the increased splenic and hepatic bacterial load resulted from an intrinsic defect in innate cell function, IFN-γ-mediated immunity, and disorganized granuloma formation. We further show that NK and NKT cells play an important role in mediating control of Salmonella in Stat4(Ity14/Ity14) mice. Stat4(Ity14/Ity14) mice had increased expression of genes involved in cell-cell interactions and communication, as well as increased CD11b expression on a subset of splenic myeloid dendritic cells, resulting in compromised recruitment of inflammatory cells to the spleen during Salmonella infection. Stat4(Ity14/Ity14) presented upregulated compensatory mechanisms, although inefficient and ultimately Stat4(Ity14/Ity14) mice develop fatal bacteremia. The following study further elucidates the pathophysiological impact of STAT4 during Salmonella infection.


Subject(s)
Gene Expression Regulation , Genetic Predisposition to Disease , Interferon-gamma/immunology , Mutation , STAT4 Transcription Factor/genetics , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/immunology , Transcription, Genetic , Animals , Bacterial Load , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cation Transport Proteins/genetics , Cluster Analysis , DNA Mutational Analysis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Loci , Immunity, Innate/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver/immunology , Liver/metabolism , Liver/microbiology , Mice , Mutation/drug effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Nitrosourea Compounds/toxicity , Pedigree , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/mortality , Salmonella typhimurium/immunology , Spleen/immunology , Spleen/metabolism , Spleen/microbiology , Transcriptome
18.
Nat Genet ; 39(3): 338-46, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17259984

ABSTRACT

We investigated the role of protein tyrosine phosphatase 1B (PTP1B) in mammary tumorigenesis using both genetic and pharmacological approaches. It has been previously shown that transgenic mice with a deletion mutation in the region of Erbb2 encoding its extracellular domain (referred to as NDL2 mice, for 'Neu deletion in extracellular domain 2') develop mammary tumors that progress to lung metastasis. However, deletion of PTP1B activity in the NDL2 transgenic mice either by breeding with Ptpn1-deficient mice or by treatment with a specific PTP1B inhibitor results in significant mammary tumor latency and resistance to lung metastasis. In contrast, specific overexpression of PTP1B in the mammary gland leads to spontaneous breast cancer development. The regulation of ErbB2-induced mammary tumorigenesis by PTB1B occurs through the attenuation of both the MAP kinase (MAPK) and Akt pathways. This report provides a rationale for the development of PTP1B as a new therapeutic target in breast cancer.


Subject(s)
Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/enzymology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Animals , Apoptosis , Cell Line, Tumor , Down-Regulation , Female , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , MAP Kinase Signaling System/physiology , Mammary Glands, Animal/drug effects , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction
19.
Hum Mol Genet ; 22(12): 2400-10, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23428429

ABSTRACT

Loss of the tumor suppressor PTEN is a common occurrence in prostate cancer. This aberration leads to the ectopic activation of the PI3K-Akt pathway, which promotes tumor growth. Here, we show that the transcription factor Gata3 is progressively lost in Pten-deficient mouse prostate tumors as a result of both transcriptional down-regulation and increased proteasomal degradation. To determine the significance of this loss, we used conditional loss- and gain-of-function approaches to manipulate Gata3 expression levels in prostate tumors. Our results show that Gata3 inactivation in Pten-deficient prostates accelerates tumor invasion. Conversely, enforced expression of GATA3 in Pten-deficient tissues markedly delays tumor progression. In Pten-deficient prostatic ducts, enforced GATA3 prevented Akt activation, which correlated with the down-regulation of Pik3cg and Pik3c2a mRNAs, encoding respectively class I and II PI3K subunits. Remarkably, the majority of human prostate tumors similarly show loss of active GATA3 as they progress to the aggressive castrate-resistant stage. In addition, GATA3 expression levels in hormone-sensitive tumors holds predictive value for tumor recurrence. Together, these data establish Gata3 as an important regulator of prostate cancer progression.


Subject(s)
GATA3 Transcription Factor/metabolism , PTEN Phosphohydrolase/deficiency , Prostate/metabolism , Prostatic Neoplasms/metabolism , Animals , Down-Regulation , Female , GATA3 Transcription Factor/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tumor Cells, Cultured
20.
Biol Reprod ; 93(1): 16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26040673

ABSTRACT

The luteinizing hormone receptor, LHCGR, is essential for fertility in males and females, and genetic mutations in the receptor have been identified that result in developmental and reproductive defects. We have previously generated and characterized a mouse model (KiLHR(D582G)) for familial male-limited precocious puberty caused by an activating mutation in the receptor. We demonstrated that the phenotype of the KiLHR(D582G) male mice is an accurate phenocopy of male patients with activating LHCGR mutations. In this study, we observed that unlike women with activating LHCGR mutations who are normal, female KiLHR(D582G) mice are infertile. Mice exhibit irregular estrous cyclicity, anovulation, and precocious puberty. A temporal study from 2-24 wk of age indicated elevated levels of progesterone, androstenedione, testosterone, and estradiol and upregulation of several steroidogenic enzyme genes. Ovaries of KiLHR(D582G) mice exhibited significant pathology with the development of large hemorrhagic cysts as early as 3 wk of age, extensive stromal cell hyperplasia and hypertrophy with luteinization, numerous atretic follicles, and granulosa cell tumors. Ovulation could not be rescued by the addition of exogenous gonadotropins. The body weights of the KiLHR(D582G) mice were higher than wild-type counterparts, but there was no increase in the body fat composition or metabolic abnormalities such as impaired glucose tolerance and insulin resistance. These studies demonstrate that activating LHCGR mutations do not produce the same phenotype in female mice as in humans and clearly illustrate species differences in the expression and regulation of LHCGR in the ovary, but not in the testis.


Subject(s)
Anovulation/genetics , Estrous Cycle/genetics , Infertility, Female/genetics , Polycystic Ovary Syndrome/genetics , Receptors, LH/genetics , Androstenedione/blood , Animals , Anovulation/metabolism , Anovulation/pathology , Blood Glucose , Body Composition/genetics , Estradiol/blood , Estrous Cycle/metabolism , Female , Infertility, Female/metabolism , Infertility, Female/pathology , Insulin/blood , Mice , Mice, Transgenic , Mutation , Ovary/metabolism , Ovary/pathology , Polycystic Ovary Syndrome/pathology , Progesterone/blood , Receptors, LH/metabolism , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL