Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Sci Rep ; 13(1): 18227, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37880276

ABSTRACT

MED19, a component of the mediator complex and a co-regulator of the androgen receptor (AR), is pivotal in prostate cancer cell proliferation. MED19 has two isoforms: a full-length "canonical" and a shorter "alternative" variant. Specific antibodies were developed to investigate these isoforms. Both exhibit similar expression in normal prostate development and adult prostate tissue, but the canonical isoform is elevated in prostate adenocarcinomas. Overexpression of canonical MED19 in LNCaP cells promotes growth under conditions of androgen deprivation in vitro and in vivo, mirroring earlier findings with alternative MED19-overexpressing LNCaP cells. Interestingly, alternative MED19 cells displayed strong colony formation in clonogenic assays under conditions of androgen deprivation, while canonical MED19 cells did not, suggesting distinct functional roles. These isoforms also modulated gene expression differently. Canonical MED19 triggered genes related to extracellular matrix remodeling while suppressing those involved in androgen-inactivating glucuronidation. In contrast, alternative MED19 elevated genes tied to cell movement and reduced those associated with cell adhesion and differentiation. The ratio of MED19 isoform expression in prostate cancers shifts with the disease stage. Early-stage cancers exhibit higher canonical MED19 expression than alternative MED19, consistent with canonical MED19's ability to promote cell proliferation under androgen deprivation. Conversely, alternative MED19 levels were higher in later-stage metastatic prostate cancer than in canonical MED19, reflecting alternative MED19's capability to enhance cell migration and autonomous cell growth. Our findings suggest that MED19 isoforms play unique roles in prostate cancer progression and highlights MED19 as a potential therapeutic target for both early and late-stage prostate cancer.


Subject(s)
Androgens , Mediator Complex , Prostatic Neoplasms , Humans , Male , Androgens/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Mediator Complex/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
2.
Cancer Res ; 82(4): 571-585, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34903602

ABSTRACT

Pyruvate kinase M2 (PKM2) has been shown to promote tumorigenesis by facilitating the Warburg effect and enhancing the activities of oncoproteins. However, this paradigm has recently been challenged by studies in which the absence of PKM2 failed to inhibit and instead accelerated tumorigenesis in mouse models. These results seem inconsistent with the fact that most human tumors overexpress PKM2. To further elucidate the role of PKM2 in tumorigenesis, we investigated the effect of PKM2 knockout in oncogenic HRAS-driven urothelial carcinoma. While PKM2 ablation in mouse urothelial cells did not affect tumor initiation, it impaired the growth and maintenance of HRAS-driven tumors. Chemical inhibition of PKM2 recapitulated these effects. Both conditions substantially reduced complex formation of PKM2 with STAT3, their nuclear translocation, and HIF1α- and VEGF-related angiogenesis. The reduction in nuclear STAT3 in the absence of PKM2 also correlated with decreased autophagy and increased apoptosis. Time-controlled, inducible PKM2 overexpression in simple urothelial hyperplasia did not trigger tumorigenesis, while overexpression of PKM2, but not PKM1, in nodular urothelial hyperplasia with angiogenesis strongly accelerated tumorigenesis. Finally, in human patients, PKM2 was overexpressed in low-grade nonmuscle-invasive and high-grade muscle-invasive bladder cancer. Based on these data, PKM2 is not required for tumor initiation but is essential for tumor growth and maintenance by enhancing angiogenesis and metabolic addiction. The PKM2-STAT3-HIF1α/VEGF signaling axis may play a critical role in bladder cancer and may serve as an actionable therapeutic target. SIGNIFICANCE: Genetic manipulation and pharmacologic inhibition of PKM2 in mouse urothelial lesions highlight its essential role in promoting angiogenesis and metabolic addiction, events indispensable for tumor growth and maintenance.


Subject(s)
Carcinoma, Transitional Cell/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Pyruvate Kinase/genetics , Urinary Bladder Neoplasms/genetics , Active Transport, Cell Nucleus/genetics , Animals , Apoptosis/genetics , Autophagy/genetics , Carcinogenesis/genetics , Carcinoma, Transitional Cell/blood supply , Carcinoma, Transitional Cell/metabolism , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pyruvate Kinase/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Mol Biol Cell ; 29(26): 3128-3143, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30303751

ABSTRACT

Uroplakin (UP) tetraspanins and their associated proteins are major mammalian urothelial differentiation products that form unique two-dimensional crystals of 16-nm particles ("urothelial plaques") covering the apical urothelial surface. Although uroplakins are highly expressed only in mammalian urothelium and are often referred to as being urothelium specific, they are also expressed in several mouse nonurothelial cell types in stomach, kidney, prostate, epididymis, testis/sperms, and ovary/oocytes. In oocytes, uroplakins colocalize with CD9 on cell-surface and multivesicular body-derived exosomes, and the cytoplasmic tail of UPIIIa undergoes a conserved fertilization-dependent, Fyn-mediated tyrosine phosphorylation that also occurs in Xenopus laevis eggs. Uroplakin knockout and antibody blocking reduce mouse eggs' fertilization rate in in vitro fertilization assays, and UPII/IIIa double-knockout mice have a smaller litter size. Phylogenetic analyses showed that uroplakin sequences underwent significant mammal-specific changes. These results suggest that, by mediating signal transduction and modulating membrane stability that do not require two-dimensional-crystal formation, uroplakins can perform conserved and more ancestral fertilization functions in mouse and frog eggs. Uroplakins acquired the ability to form two-dimensional-crystalline plaques during mammalian divergence, enabling them to perform additional functions, including umbrella cell enlargement and the formation of permeability and mechanical barriers, to protect/modify the apical surface of the modern-day mammalian urothelium.


Subject(s)
Genetic Speciation , Oocytes/metabolism , Ovary/metabolism , Uroplakins/genetics , Urothelium/metabolism , Zygote/metabolism , Animals , Cell Differentiation , Female , Fertilization/genetics , Gene Expression Regulation , Litter Size , Male , Mice , Mice, Knockout , Oocytes/cytology , Ovary/cytology , Parthenogenesis/genetics , Phosphorylation , Phylogeny , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Signal Transduction , Testis/cytology , Testis/metabolism , Tetraspanin 29/genetics , Tetraspanin 29/metabolism , Uroplakins/classification , Uroplakins/metabolism , Urothelium/cytology , Xenopus laevis , Zygote/cytology
12.
Am J Pathol ; 184(10): 2598-607, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25088980

ABSTRACT

The androgen receptor (AR) in stromal cells contributes significantly to the development and growth of prostate during fetal stages as well as during prostate carcinogenesis and cancer progression. During prostate development, stromal AR induces and promotes epithelial cell growth, as observed from tissue recombinant and mouse knockout studies. During prostate carcinogenesis and progression, the stromal cells begin to lose AR expression as early as at the stage of high-grade prostatic intraepithelial neoplasia. The extent of loss of stromal AR is directly proportional to the degree of differentiation (Gleason grade) and progression of prostate cancer (PCa). Co-culture studies suggested that stromal AR inhibits the growth of malignant epithelial cells, possibly through expression of certain paracrine factors in the presence of androgens. This functional reversal of stromal AR, from growth promotion during fetal prostate development to mediating certain growth-inhibiting effects in cancer, explains to some extent the reason that loss of AR expression in stromal cells may be crucial for development of resistance to androgen ablation therapy for PCa. From a translational perspective, it generates the need to re-examine the current therapeutic options and opens a fundamental new direction for therapeutic interventions, especially in advanced PCa.


Subject(s)
Androgens/metabolism , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Male , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology
13.
Clin Perinatol ; 41(3): 679-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25155735

ABSTRACT

Congenital anomalies of the upper urinary tract are common and frequently diagnosed on prenatal ultrasound. In the absence of infection, these anomalies are often asymptomatic. This article reviews key features and long-term implications to assist in discussions with families. In contrast, a perinatal renal tumor is rare but extremely alarming. This update on the most common tumors and their treatment is useful in reassuring parents that most infants, after primary surgical resection, are cured without adjuvant therapies. To understand renal agenesis and other congenital renal malformations and their associated anomalies, a brief review of normal renal development is presented.


Subject(s)
Kidney Neoplasms , Urinary Tract/abnormalities , Urogenital Abnormalities/diagnosis , Female , Global Health , Humans , Incidence , Infant, Newborn , Kidney Neoplasms/diagnosis , Kidney Neoplasms/embryology , Kidney Neoplasms/epidemiology , Pregnancy , Prenatal Diagnosis , Urogenital Abnormalities/epidemiology
14.
Biochim Biophys Acta ; 1842(2): 154-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24263171

ABSTRACT

The Cancer/Testis Antigen (CTA), Prostate-associated Gene 4 (PAGE4), is a stress-response protein that is upregulated in prostate cancer (PCa) especially in precursor lesions that result from inflammatory stress. In cells under stress, translocation of PAGE4 to mitochondria increases while production of reactive oxygen species decreases. Furthermore, PAGE4 is also upregulated in human fetal prostate, underscoring its potential role in development. However, the proteins that interact with PAGE4 and the mechanisms underlying its pleiotropic functions in prostatic development and disease remain unknown. Here, we identified c-Jun as a PAGE4 interacting partner. We show that both PAGE4 and c-Jun are overexpressed in the human fetal prostate; and in cell-based assays, PAGE4 robustly potentiates c-Jun transactivation. Single-molecule Förster resonance energy transfer experiments indicate that upon binding to c-Jun, PAGE4 undergoes conformational changes. However, no interaction is observed in presence of BSA or unilamellar vesicles containing the mitochondrial inner membrane diphosphatidylglycerol lipid marker cardiolipin. Together, our data indicate that PAGE4 specifically interacts with c-Jun and that, conformational dynamics may account for its observed pleiotropic functions. To our knowledge, this is the first report demonstrating crosstalk between a CTA and a proto-oncogene. Disrupting PAGE4/c-Jun interactions using small molecules may represent a novel therapeutic strategy for PCa.


Subject(s)
Antigens, Neoplasm/metabolism , Prostate/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcriptional Activation , 3' Untranslated Regions/genetics , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Prostate/embryology , Prostate/growth & development , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Protein Conformation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-jun/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological
16.
Rev Urol ; 14(1-2): 36-8, 2012.
Article in English | MEDLINE | ID: mdl-23173000
17.
Rev Urol ; 14(1-2): 38, 2012.
Article in English | MEDLINE | ID: mdl-23173001
18.
Carcinogenesis ; 33(4): 770-80, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22287562

ABSTRACT

Although formation of urothelial carcinoma of the bladder (UCB) requires multiple steps and proceeds along divergent pathways, the underlying genetic and molecular determinants for each step and pathway remain undefined. By developing transgenic mice expressing single or combinatorial genetic alterations in urothelium, we demonstrated here that overcoming oncogene-induced compensatory tumor barriers was critical for urothelial tumor initiation. Constitutively active Ha-ras (Ras*) elicited urothelial hyperplasia that was persistent and did not progress to tumors over a 10 months period. This resistance to tumorigenesis coincided with increased expression of p53 and all pRb family proteins. Expression of a Simian virus 40 T antigen (SV40T), which disables p53 and pRb family proteins, in urothelial cells expressing Ras* triggered early-onset, rapidly-growing and high-grade papillary UCB that strongly resembled the human counterpart (pTaG3). Urothelial cells expressing both Ras* and SV40T had defective G(1)/S checkpoint, elevated Ras-GTPase and hyperactivated AKT-mTOR signaling. Inhibition of the AKT-mTOR pathway with rapamycin significantly reduced the size of high-grade papillary UCB but hyperactivated mitogen-activated protein kinase (MAPK). Inhibition of AKT-mTOR, MAPK and STAT3 altogether resulted in much greater tumor reduction and longer survival than did inhibition of AKT-mTOR pathway alone. Our studies provide the first experimental evidence delineating the combinatorial genetic events required for initiating high-grade papillary UCB, a poorly defined and highly challenging clinical entity. Furthermore, they suggest that targeted therapy using a single agent such as rapamycin may not be highly effective in controlling high-grade UCB and that combination therapy employing inhibitors against multiple targets are more likely to achieve desirable therapeutic outcomes.


Subject(s)
Signal Transduction , Urinary Bladder Neoplasms/pathology , Animals , Base Sequence , Blotting, Western , DNA Primers , GTP Phosphohydrolases/metabolism , Humans , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism
19.
PLoS One ; 7(1): e29372, 2012.
Article in English | MEDLINE | ID: mdl-22253716

ABSTRACT

BACKGROUND: During normal development in human and other placental mammals, the embryonic cloacal cavity separates along the axial longitudinal plane to give rise to the urethral system, ventrally, and the rectum, dorsally. Defects in cloacal development are very common and present clinically as a rectourethral fistula in about 1 in 5,000 live human births. Yet, the cellular mechanisms of cloacal septation remain poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: We previously detected Bone morphogenetic protein 7 (Bmp7) expression in the urorectal mesenchyme (URM), and have shown that loss of Bmp7 function results in the arrest of cloacal septation. Here, we present evidence that cloacal partitioning is driven by Bmp7 signaling in the cloacal endoderm. We performed TUNEL and immunofluorescent analysis on cloacal sections from Bmp7 null and control littermate embryos. We found that loss of Bmp7 results in a dramatic decrease in the endoderm survival and a delay in differentiation. We used immunological methods to show that Bmp7 functions by activating the c-Jun N-terminal kinase (JNK) pathway. We carried out confocal and 3D imaging analysis of mitotic chromosome bundles to show that during normal septation cells in the cloacal endoderm divide predominantly in the apical-basal direction. Loss of Bmp7/JNK signaling results in randomization of mitotic angles in the cloacal endoderm. We also conducted immunohistochemical analysis of human fetal sections to show that BMP/phospho-SMAD and JNK pathways function in the human cloacal region similar as in the mouse. CONCLUSION/SIGNIFICANCE: Our results strongly indicate that Bmp7/JNK signaling regulates remodeling of the cloacal endoderm resulting in a topological separation of the urinary and digestive systems. Our study points to the importance of Bmp and JNK signaling in cloacal development and rectourethral malformations.


Subject(s)
Body Patterning , Bone Morphogenetic Protein 7/metabolism , Cell Polarity , Cloaca/cytology , Cloaca/embryology , Animals , Bone Morphogenetic Protein 7/deficiency , Cell Differentiation , Cell Division , Cell Proliferation , Cell Survival , Cloaca/enzymology , Endoderm/cytology , Epithelium/embryology , Fetus/embryology , Fetus/enzymology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Mesoderm/cytology , Mesoderm/metabolism , Mice , Models, Biological , Rectum/embryology , Rectum/enzymology , Smad Proteins/metabolism , Urethra/embryology , Urethra/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...