Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Nat Commun ; 13(1): 6041, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36253360

ABSTRACT

Tumors exhibit enhancer reprogramming compared to normal tissue. The etiology is largely attributed to cell-intrinsic genomic alterations. Here, using freshly resected primary CRC tumors and patient-matched adjacent normal colon, we find divergent epigenetic landscapes between CRC tumors and cell lines. Intriguingly, this phenomenon extends to highly recurrent aberrant super-enhancers gained in CRC over normal. We find one such super-enhancer activated in epithelial cancer cells due to surrounding inflammation in the tumor microenvironment. We restore this super-enhancer and its expressed gene, PDZK1IP1, following treatment with cytokines or xenotransplantation into nude mice, thus demonstrating cell-extrinsic etiology. We demonstrate mechanistically that PDZK1IP1 enhances the reductive capacity CRC cancer cells via the pentose phosphate pathway. We show this activation enables efficient growth under oxidative conditions, challenging the previous notion that PDZK1IP1 acts as a tumor suppressor in CRC. Collectively, these observations highlight the significance of epigenomic profiling on primary specimens.


Subject(s)
Colorectal Neoplasms , Tumor Microenvironment , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Cytokines/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic , Mice , Mice, Nude , Tumor Microenvironment/genetics
3.
FEBS J ; 287(11): 2201-2211, 2020 06.
Article in English | MEDLINE | ID: mdl-32147971

ABSTRACT

In June of 2019, the International Cell Death Society (ICDS) held its 25th anniversary meeting in New York City at the Icahn School of Medicine at Mount Sinai organized by Drs. Richard A. Lockshin (St. John's University, USA), Zahra Zakeri (Queens College, USA), and Jerry Edward Chipuk (Icahn School of Medicine at Mount Sinai, USA). The three-day event, entitled 'Cell death through the ages: The ICDS 25th anniversary meeting', hosted ninety-one delegates including thirty-four speakers and twenty-two poster presentations. Additionally, the organizers gave special recognition to the twenty-one previous ICDS Lifetime Achievement awardees-those who have significantly contributed to the field of cell death and the growth of the organization. Here, we provide a summary of the meeting and highlight trending research in the fields of cell death, autophagy, immunology, and their impact on health and disease.


Subject(s)
Anniversaries and Special Events , Cell Death/genetics , Humans , New York City
4.
Mol Cell ; 74(3): 452-465.e7, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30879903

ABSTRACT

Signaling diversity and subsequent complexity in higher eukaryotes is partially explained by one gene encoding a polypeptide with multiple biochemical functions in different cellular contexts. For example, mouse double minute 2 (MDM2) is functionally characterized as both an oncogene and a tumor suppressor, yet this dual classification confounds the cell biology and clinical literatures. Identified via complementary biochemical, organellar, and cellular approaches, we report that MDM2 negatively regulates NADH:ubiquinone oxidoreductase 75 kDa Fe-S protein 1 (NDUFS1), leading to decreased mitochondrial respiration, marked oxidative stress, and commitment to the mitochondrial pathway of apoptosis. MDM2 directly binds and sequesters NDUFS1, preventing its mitochondrial localization and ultimately causing complex I and supercomplex destabilization and inefficiency of oxidative phosphorylation. The MDM2 amino-terminal region is sufficient to bind NDUFS1, alter supercomplex assembly, and induce apoptosis. Finally, this pathway is independent of p53, and several mitochondrial phenotypes are observed in Drosophila and murine models expressing transgenic Mdm2.


Subject(s)
Mitochondria/metabolism , NADH Dehydrogenase/genetics , Oxidative Stress/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics , A549 Cells , Animals , Apoptosis/genetics , Cell Respiration/genetics , Cytosol/metabolism , Drosophila melanogaster/genetics , Electron Transport Complex I/genetics , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Signal Transduction/genetics
5.
J Biol Chem ; 292(28): 11727-11739, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28546431

ABSTRACT

The mitochondrial network is a major site of ATP production through the coupled integration of the electron transport chain (ETC) with oxidative phosphorylation. In melanoma arising from the V600E mutation in the kinase v-RAF murine sarcoma viral oncogene homolog B (BRAFV600E), oncogenic signaling enhances glucose-dependent metabolism while reducing mitochondrial ATP production. Likewise, when BRAFV600E is pharmacologically inhibited by targeted therapies (e.g. PLX-4032/vemurafenib), glucose metabolism is reduced, and cells increase mitochondrial ATP production to sustain survival. Therefore, collateral inhibition of oncogenic signaling and mitochondrial respiration may help enhance the therapeutic benefit of targeted therapies. Honokiol (HKL) is a well tolerated small molecule that disrupts mitochondrial function; however, its underlying mechanisms and potential utility with targeted anticancer therapies remain unknown. Using wild-type BRAF and BRAFV600E melanoma model systems, we demonstrate here that HKL administration rapidly reduces mitochondrial respiration by broadly inhibiting ETC complexes I, II, and V, resulting in decreased ATP levels. The subsequent energetic crisis induced two cellular responses involving cyclin-dependent kinases (CDKs). First, loss of CDK1-mediated phosphorylation of the mitochondrial division GTPase dynamin-related protein 1 promoted mitochondrial fusion, thus coupling mitochondrial energetic status and morphology. Second, HKL decreased CDK2 activity, leading to G1 cell cycle arrest. Importantly, although pharmacological inhibition of oncogenic MAPK signaling increased ETC activity, co-treatment with HKL ablated this response and vastly enhanced the rate of apoptosis. Collectively, these findings integrate HKL action with mitochondrial respiration and shape and substantiate a pro-survival role of mitochondrial function in melanoma cells after oncogenic MAPK inhibition.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Complex II/antagonists & inhibitors , Electron Transport Complex I/antagonists & inhibitors , Lignans/pharmacology , Mitochondria/drug effects , Protein Kinase Inhibitors/pharmacology , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , G1 Phase/drug effects , Humans , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/enzymology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oxidative Phosphorylation/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Uncoupling Agents/pharmacology
6.
Prostate ; 77(2): 185-195, 2017 02.
Article in English | MEDLINE | ID: mdl-27718274

ABSTRACT

BACKGROUND: Improving our ability to predict cancer progression and response to conservative or radical intent therapy is critical if we are to prevent under or over treatment of individual patients. Whereas the majority of solid tumors now have a range of molecular and/or immunological markers to help define prognosis and treatment options, prostate cancer still relies mainly on histological grading and clinical parameters. We have recently reported that androgen receptor (AR) expression in stroma inversely associates with prostate cancer-specific survival, and that stromal AR reduces metastasis. For this paper, we tested the hypothesis that the AR-regulated gene FKBP51 could be used as a marker of AR activity to better predict outcome. METHODS: Using immunohistochemistry on a cohort of 64 patient-matched benign and malignant prostate tissues, we assessed patient outcome by FKBP51 and AR levels. Immunoblot and RT-qPCR were used to demonstrate androgen regulation of FKBP51 in primary and primary human prostatic fibroblasts and fibroblast cell-lines. RESULTS: As predicted by FKBP51 level, high AR activity in cancer stroma was associated with longer median survival (1,306 days) compared with high AR alone (699 days), whereas those with low AR and/or low FKBP51 did poorly (384 and 338 days, respectively). Survival could not be predicted on the basis cancer epithelial AR levels or activity, and was not associated with immunoreactivity in patient matched benign tissues. CONCLUSION: FKBP51 improves the ability of stromal AR to predict prostate cancer-specific mortality. By adding additional immunological assessment, similar to what is already in place in a number of other cancers, we could better serve patients with prostate cancer in prognosis and informed treatment choices. Prostate 77:185-195, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Biomarkers, Tumor/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Tacrolimus Binding Proteins/metabolism , Cell Line, Tumor , Cohort Studies , Humans , Male , Prognosis , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Cells, Cultured
8.
BMC Cancer ; 15: 791, 2015 Oct 24.
Article in English | MEDLINE | ID: mdl-26498662

ABSTRACT

BACKGROUND: In breast cancer, progesterone receptor (PR) positivity or abundance is positively associated with survival and treatment response. It was initially believed that PR was a useful diagnostic marker of estrogen receptor activity, but increasingly PR has been recognised to play an important biological role in breast homeostasis, carcinogenesis and metastasis. Although PR expression is almost exclusively observed in estrogen receptor positive tumors, few studies have investigated the cellular mechanisms of PR action in the context of ongoing estrogen signalling. METHODS: In this study, we contrast PR function in estrogen pretreated ZR-75-1 breast cancer cells with vehicle treated ZR-75-1 and T-47D breast cancer cells using expression microarrays and chromatin immunoprecipitation-sequencing. RESULTS: Estrogen cotreatment caused a dramatic increase in the number of genes regulated by progesterone in ZR-75-1 cells. In T-47D cells that have naturally high levels of PR, estrogen and progesterone cotreatment resulted in a reduction in the number of regulated genes in comparison to treatment with either hormone alone. At a genome level, estrogen pretreatment of ZR-75-1 cells led to a 10-fold increase in the number of PR DNA binding sites detected using ChIP-sequencing. Time course assessment of progesterone regulated genes in the context of estrogen pretreatment highlighted a series of important regulatory pathways, including those driven by epithelial growth factor receptor (EGFR). Importantly, progesterone applied to cells pretreated with estradiol resulted in switching of the PAM50-determined intrinsic breast cancer subtype from Luminal A to Basal-like, and increased the Oncotype DX® Unscaled Recurrence Score. CONCLUSION: Estrogen pretreatment of breast cancer cells increases PR steady state levels, resulting in an unequivocal progesterone response that upregulates key members of growth factor pathways. The transformative changes progesterone exerts on the breast cancer subtype suggest that these subtyping tools should be used with caution in premenopausal women.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , ErbB Receptors/biosynthesis , Estrogens/administration & dosage , Progesterone/administration & dosage , Transcriptional Activation/drug effects , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Receptors, Progesterone/biosynthesis , Transcriptional Activation/physiology , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/physiology
9.
J Biol Chem ; 290(39): 24007-20, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26254468

ABSTRACT

Growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are oocyte-specific growth factors with central roles in mammalian reproduction, regulating species-specific fecundity, ovarian follicular somatic cell differentiation, and oocyte quality. In the human, GDF9 is produced in a latent form, the mechanism of activation being an open question. Here, we produced a range of recombinant GDF9 and BMP15 variants, examined their in silico and physical interactions and their effects on ovarian granulosa cells (GC) and oocytes. We found that the potent synergistic actions of GDF9 and BMP15 on GC can be attributed to the formation of a heterodimer, which we have termed cumulin. Structural modeling of cumulin revealed a dimerization interface identical to homodimeric GDF9 and BMP15, indicating likely formation of a stable complex. This was confirmed by generation of recombinant heterodimeric complexes of pro/mature domains (pro-cumulin) and covalent mature domains (cumulin). Both pro-cumulin and cumulin exhibited highly potent bioactivity on GC, activating both SMAD2/3 and SMAD1/5/8 signaling pathways and promoting proliferation and expression of a set of genes associated with oocyte-regulated GC differentiation. Cumulin was more potent than pro-cumulin, pro-GDF9, pro-BMP15, or the two combined on GC. However, on cumulus-oocyte complexes, pro-cumulin was more effective than all other growth factors at notably improving oocyte quality as assessed by subsequent day 7 embryo development. Our results support a model of activation for human GDF9 dependent on cumulin formation through heterodimerization with BMP15. Oocyte-secreted cumulin is likely to be a central regulator of fertility in mono-ovular mammals.


Subject(s)
Bone Morphogenetic Protein 15/metabolism , Granulosa Cells/metabolism , Growth Differentiation Factor 9/metabolism , Oocytes/metabolism , Animals , Bone Morphogenetic Protein 15/genetics , Female , Granulosa Cells/cytology , Growth Differentiation Factor 9/genetics , Humans , Mice , Oocytes/cytology , Protein Multimerization/physiology , Signal Transduction/physiology , Smad Proteins/genetics , Smad Proteins/metabolism
10.
Oncotarget ; 6(18): 16135-50, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-25965833

ABSTRACT

Androgen receptor (AR) signaling in stromal cells is important in prostate cancer, yet the mechanisms underpinning stromal AR contribution to disease development and progression remain unclear. Using patient-matched benign and malignant prostate samples, we show a significant association between low AR levels in cancer associated stroma and increased prostate cancer-related death at one, three and five years post-diganosis, and in tissue recombination models with primary prostate cancer cells that low stromal AR decreases castration-induced apoptosis. AR-regulation was found to be different in primary human fibroblasts isolated from adjacent to cancerous and non-cancerous prostate epithelia, and to represent altered activation of myofibroblast pathways involved in cell cycle, adhesion, migration, and the extracellular matrix (ECM). Without AR signaling, the fibroblast-derived ECM loses the capacity to promote attachment of both myofibroblasts and cancer cells, is less able to prevent cell-matrix disruption, and is less likely to impede cancer cell invasion. AR signaling in prostate cancer stroma appears therefore to alter patient outcome by maintaining an ECM microenvironment inhibitory to cancer cell invasion. This paper provides comprehensive insight into AR signaling in the non-epithelial prostate microenvironment, and a resource from which the prognostic and therapeutic implications of stromal AR levels can be further explored.


Subject(s)
Myofibroblasts/pathology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Stromal Cells/pathology , Tumor Microenvironment , Aged , Aged, 80 and over , Androgens/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Case-Control Studies , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Follow-Up Studies , Humans , Immunoenzyme Techniques , Male , Middle Aged , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Neoplasm Grading , Neoplasm Invasiveness , Orchiectomy , Prognosis , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/metabolism , Tissue Array Analysis , Tumor Cells, Cultured
11.
Mol Cell Endocrinol ; 384(1-2): 185-99, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24440747

ABSTRACT

There is extensive knowledge of androgen receptor (AR) signaling in cancer cells, but less regarding androgen action in stromal cells of the tumor microenvironment. We report here the genome-wide effects of a stromal cell specific molecular adapter and AR coregulator, hydrogen peroxide-inducible gene 5 (Hic-5/TGFB1I1), on AR function in prostate myofibroblasts. Following androgen stimulation, Hic-5 rapidly translocates to the nucleus, coincident with increased phosphorylation of focal adhesion kinase. As a coregulator, Hic-5 acted to amplify or inhibit regulation of approximately 50% of AR target genes, affected androgen regulation of growth, cell adhesion, motility and invasion. These data suggest Hic-5 as a transferable adaptor between focal adhesions and the nucleus of prostate myofibroblasts, where it acts a key mediator of the specificity and sensitivity of AR signaling. We propose a model in which Hic-5 coordinates AR signaling with adhesion and extracellular matrix contacts to regulate cell behavior in the tumor microenvironment.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/genetics , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Myofibroblasts/metabolism , Receptors, Androgen/genetics , Stromal Cells/metabolism , Androgens/pharmacology , Cell Adhesion , Cell Line, Transformed , Cell Movement , Cell Nucleus/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Male , Molecular Sequence Annotation , Myofibroblasts/cytology , Myofibroblasts/drug effects , Phosphorylation , Prostate/metabolism , Prostate/pathology , Protein Transport , Receptors, Androgen/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/drug effects , Testosterone/pharmacology , Tumor Microenvironment
12.
Mol Cell Endocrinol ; 382(2): 899-908, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24239616

ABSTRACT

Medroxyprogesterone acetate (MPA) has widely been used in hormone replacement therapy (HRT), and is associated with an increased risk of breast cancer, possibly due to disruption of androgen receptor (AR) signaling. In contrast, the synthetic HRT Tibolone does not increase breast density, and is rapidly metabolized to estrogenic 3α-OH-tibolone and 3ß-OH-tibolone, and a delta-4 isomer (Δ(4)-TIB) that has both androgenic and progestagenic properties. Here, we show that 5α-dihydrotestosterone (DHT) and Δ(4)-TIB, but not MPA, stabilize AR protein levels, initiate specific AR intramolecular interactions critical for AR transcriptional regulation, and increase proliferation of AR positive MDA-MB-453 breast cancer cells. Structural modeling and molecular dynamic simulation indicate that Δ(4)-TIB induces a more stable AR structure than does DHT, and MPA a less stable one. Microarray expression analyses confirms that the molecular actions of Δ(4)-TIB more closely resembles DHT in breast cancer cells than either ligand does to MPA.


Subject(s)
Androgens/pharmacology , Dihydrotestosterone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/genetics , Norpregnenes/pharmacology , Receptors, Androgen/genetics , Androgens/chemistry , Androgens/metabolism , Biotransformation , Cell Line, Tumor , Dihydrotestosterone/chemistry , Dihydrotestosterone/metabolism , Female , Gene Expression Profiling , Humans , Medroxyprogesterone Acetate/chemistry , Medroxyprogesterone Acetate/pharmacology , Molecular Dynamics Simulation , Neoplasm Proteins/metabolism , Norpregnanes/metabolism , Norpregnenes/chemistry , Norpregnenes/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Androgen/metabolism , Structure-Activity Relationship
13.
Int J Cancer ; 133(12): 2812-23, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23740762

ABSTRACT

Solid tumors have an increased reliance on Hsp70/Hsp90 molecular chaperones for proliferation, survival and maintenance of intracellular signaling systems. An underinvestigated component of the chaperone system is the tetratricopeptide repeat (TPR)-containing cochaperone, which coordinates Hsp70/Hsp90 involvement on client proteins as well as having diverse individual actions. A potentially important cochaperone in prostate cancer (PCa) is small glutamine-rich TPR-containing protein alpha (SGTA), which interacts with the androgen receptor (AR) and other critical cancer-related client proteins. In this study, the authors used small interfering RNA coupled with genome-wide expression profiling to investigate the biological significance of SGTA in PCa and its influence on AR signaling. Knockdown of SGTA for 72 hr in PCa C4-2B cells significantly altered expression of >1,900 genes (58% decreased) and reduced cell proliferation (p < 0.05). The regulation of 35% of 5α-dihydrotestosterone (DHT) target genes was affected by SGTA knockdown, with gene-specific effects on basal or DHT-induced expression or both. Pathway analysis revealed a role for SGTA in p53, generic PCa and phosphoinositol kinase (PI3K) signaling pathways; the latter evident by a reduction in PI3K subunit p100ß levels and decreased phosphorylated Akt. Immunohistochemical analysis of 64 primary advanced PCa samples showed a significant increase in the AR:SGTA ratio in cancerous lesions compared to patient-matched benign prostatic hyperplasia tissue (p < 0.02). This study not only provides insight into the biological actions of SGTA and its effect on genome-wide AR transcriptional activity and other therapeutically targeted intracellular signaling pathways but also provides evidence for PCa-specific alterations in SGTA expression.


Subject(s)
Carrier Proteins/physiology , Cell Proliferation , Phosphatidylinositol 3-Kinases/physiology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/physiology , Receptors, Androgen/physiology , Signal Transduction/physiology , Animals , Carrier Proteins/analysis , Carrier Proteins/antagonists & inhibitors , Dihydrotestosterone/pharmacology , Humans , Male , Mice , Molecular Chaperones , Prostatic Neoplasms/metabolism , Receptors, Androgen/analysis , Transcriptome , Tumor Suppressor Protein p53/analysis
14.
J Mol Endocrinol ; 49(2): 57-68, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22693264

ABSTRACT

Ligand-dependent activity of steroid receptors is affected by tetratricopeptide repeat (TPR)-containing co-chaperones, such as small glutamine-rich tetratricopeptide repeat-containing alpha (SGTA). However, the precise mechanisms by which the predominantly cytoplasmic TPR proteins affect downstream transcriptional outcomes of steroid signaling remain unclear. In this study, we assessed how SGTA affects ligand sensitivity and action of the androgen receptor (AR) using a transactivation profiling approach. Deletion mapping coupled with structural prediction, transcriptional assays, and in vivo regulation of AR-responsive promoters were used to assess the role of SGTA domains in AR responses. At subsaturating ligand concentrations of ≤ 0.1 nM 5α-dihydrotestosterone, SGTA overexpression constricted AR activity by an average of 32% (P<0.002) across the majority of androgen-responsive loci tested, as well as on endogenous promoters in vivo. The strength of the SGTA effect was associated with the presence or absence of bioinformatically predicated transcription factor motifs at each site. Homodimerizaion of SGTA, which is thought to be necessary for chaperone complex formation, was found to be dependent on the structural integrity of amino acids 1-80, and a core evolutionary conserved peptide within this region (amino acids 21-40) necessary for an effect of SGTA on the activity of both exogenous and endogenous AR. This study provides new insights into the subdomain structure of SGTA and how SGTA acts as a regulator of AR ligand sensitivity. A change in AR:SGTA ratio will impact the cellular and molecular response of prostate cancer cells to maintain androgenic signals, which may influence tumor progression.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Molecular Chaperones/metabolism , Receptors, Androgen/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins/genetics , Chlorocebus aethiops , Conserved Sequence , Dihydrotestosterone/metabolism , Genes, Reporter , Ligands , Protein Multimerization , Protein Structure, Tertiary , Receptors, Androgen/genetics , Transcription Factors/metabolism
15.
J Pediatr Gastroenterol Nutr ; 55(1): 26-31, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22193181

ABSTRACT

OBJECTIVES: Intestinal crypt fission peaks during infancy. In human and experimental familial polyposis coli, increased crypt fission is due to activation of Wnt/ß-catenin signalling, but the molecular basis of crypt fission during intestinal growth has not been examined. The aim of this project was to investigate whether crypt fission and intestinal growth are affected by experimental blockade of the Wnt/ß-catenin signalling pathway. METHODS: Hooded Wistar rats were given either the Wnt inhibitor, dickkopf (30 and 100 ng), daily or vehicle control intraperitoneally from days 11 to 15 and were killed at day 16. Intestinal morphometry was used to measure villous area, crypt area, percentage of crypt fission, and crypt mitotic count. Intestinal stem cells were assessed by expression of real time-polymerase chain reaction for Lgr5 (a stem cell marker), and the number of ß-catenin-expressing crypts by immunostaining was determined after 100-ng dickkopf treatment. RESULTS: Dickkopf at 30 and 100 ng/day reduced villous area to 71% (P = 0.013) and 29% (P < 0.0001), crypt area to 42% (P = 0.0026) and 30% (P = 0.0067), and crypt fission to 51% (P = 0.006) and 29% (P < 0.0001), respectively, of control values. Mitotic count per crypt did not change. Lgr5 RNA expression and the number of ß-catenin-expressing crypts decreased in dickkopf-treated animals. CONCLUSIONS: We conclude that intestinal crypt fission during infancy is mediated by Wnt signalling. It is possible that local treatment with Wnt agonists could be used to increase intestinal growth.


Subject(s)
Intercellular Signaling Peptides and Proteins/pharmacology , Intestinal Mucosa/metabolism , Intestines/drug effects , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Intestines/pathology , Mitotic Index , Polycomb Repressive Complex 1/drug effects , Polycomb Repressive Complex 1/metabolism , RNA/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , beta Catenin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...