Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 120(16): e2210418120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37040401

ABSTRACT

The hypoxia-inducible factor 1-α (HIF-1α) enables cells to adapt and respond to hypoxia (Hx), and the activity of this transcription factor is regulated by several oncogenic signals and cellular stressors. While the pathways controlling normoxic degradation of HIF-1α are well understood, the mechanisms supporting the sustained stabilization and activity of HIF-1α under Hx are less clear. We report that ABL kinase activity protects HIF-1α from proteasomal degradation during Hx. Using a fluorescence-activated cell sorting (FACS)-based CRISPR/Cas9 screen, we identified HIF-1α as a substrate of the cleavage and polyadenylation specificity factor-1 (CPSF1), an E3-ligase which targets HIF-1α for degradation in the presence of an ABL kinase inhibitor in Hx. We show that ABL kinases phosphorylate and interact with CUL4A, a cullin ring ligase adaptor, and compete with CPSF1 for CUL4A binding, leading to increased HIF-1α protein levels. Further, we identified the MYC proto-oncogene protein as a second CPSF1 substrate and show that active ABL kinase protects MYC from CPSF1-mediated degradation. These studies uncover a role for CPSF1 in cancer pathobiology as an E3-ligase antagonizing the expression of the oncogenic transcription factors, HIF-1α and MYC.


Subject(s)
Gene Expression Regulation , Transcription Factors , Humans , Cullin Proteins/metabolism , Hypoxia , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Genes, abl , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Cleavage And Polyadenylation Specificity Factor/metabolism
2.
Breast Cancer Res Treat ; 179(1): 67-77, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31562570

ABSTRACT

PURPOSE: Fulvestrant is a selective estrogen receptor downregulator (SERD) that is approved for first- or second-line use as a single agent or in combination with cyclin dependent kinase or phosphatidylinositol 3-kinase inhibitors for the treatment of metastatic breast cancer. Fulvestrant exhibits exceptionally effective antitumor activity in preclinical models of breast cancer, a success that has been attributed to its robust SERD activity despite modest receptor downregulation in patient tumors. By modeling human exposures in animal models we probe the absolute need for SERD activity. METHODS: Three xenograft models of endocrine therapy-resistant breast cancer were used to evaluate the efficacy of fulvestrant administered in doses historically used in preclinical studies in the field or by using a dose regimen intended to model clinical exposure levels. Pharmacokinetic and pharmacodynamic analyses were conducted to evaluate plasma exposure and intratumoral ER downregulation. RESULTS: A clinically relevant 25 mg/kg dose of fulvestrant exhibited antitumor efficacy comparable to the historically used 200 mg/kg dose, but at this lower dose it did not result in robust ER downregulation. Further, the antitumor efficacy of the lower dose of fulvestrant was comparable to that observed for other oral SERDs currently in development. CONCLUSION: The use of clinically unachievable exposure levels of fulvestrant as a benchmark in preclinical development of SERDs may negatively impact the selection of those molecules that are advanced for clinical development. Further, these studies suggest that antagonist efficacy, as opposed to SERD activity, is likely to be the primary driver of clinical response.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacokinetics , Breast Neoplasms/drug therapy , Estrogen Receptor Antagonists/pharmacokinetics , Fulvestrant/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Dose-Response Relationship, Drug , Estrogen Receptor Antagonists/administration & dosage , Estrogen Receptor alpha/antagonists & inhibitors , Female , Fulvestrant/administration & dosage , Mice , Xenograft Model Antitumor Assays
3.
Breast Cancer Res Treat ; 179(3): 769, 2020 02.
Article in English | MEDLINE | ID: mdl-31734822

ABSTRACT

The article Pharmacokinetic and pharmacodynamic analysis of fulvestrant in preclinical models of breast cancer to assess the importance of its estrogen receptor-α degrader activity in antitumor efficacy, written by Suzanne E. Wardell, Alexander P. Yllanes, Christina A. Chao, Yeeun Bae, Kaitlyn J. Andreano, Taylor K. Desautels, Kendall A. Heetderks, Jeremy T. Blitzer, John D. Norris, Donald P. McDonnell, was originally published electronically on the publisher's internet portal on September 27, 2019 without open access. With the author(s)' decision to opt for Open Choice the copyright of the article changed on November 16, 2019 to © The Author(s) 2019 and the article is forthwith distributed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The original article has been corrected.

4.
Prostate ; 79(4): 414-424, 2019 03.
Article in English | MEDLINE | ID: mdl-30560549

ABSTRACT

BACKGROUND: The recurrent p.Gly84Glu germline mutation (G84E) in HOXB13 is consistently associated with prostate cancer (PCa), although the mechanisms underlying such linkage remain elusive. The majority of the PCa-associated HOXB13 mutations identified are localized to two conserved domains in HOXB13 that have been shown to mediate the interaction with MEIS cofactors belonging to the TALE family of homeodomain transcription factors. In this study, we sought to interrogate the biochemical and functional interactions between HOXB13 and MEIS in prostatic cells with a goal of defining how the HOXB13-MEIS complex impacts PCa pathobiology and define the extent to which the oncogenic activity of G84E is related to its effect on HOXB13-MEIS interaction/function. METHODS: HOXB13 and MEIS paralog expression in prostate epithelial cells and PCa cell lines was characterized by qPCR and immunoblot analyses. HOXB13 and MEIS1 co-expression in human prostate tissue was confirmed by IHC, followed by co-IP mapping of HOXB13-MEIS1 interactions. Proliferation of the PCa cell line LAPC4 following shRNA-mediated knockdown of each gene or both genes was assessed using DNA- and metabolic-based assays. Transcriptional targets of HOXB13 and MEIS1 were identified by gene expression profiling and qPCR. Finally, protein stability of HOXB13 in the context of MEIS1 was determined using pulse-chase assays. RESULTS: HOXB13 and MEIS1 are co-expressed and interact in prostate cells. Both of the putative MEIS interacting domains (MID) within HOXB13 were shown to be capable of mediating the interaction between HOXB13 and MEIS1 independently and such interactions were not influenced by the G84E mutation. The inhibitory effect of either HOXB13 or MEIS1 knockdown on cellular proliferation was augmented by knockdown of both genes, and MEIS1 knockdown abolished HOXB13-driven regulation of BCHE and TNFSF10 mRNA expression. Notably, we demonstrated that MEIS1 stabilized the HOXB13 protein in LAPC4 cells. CONCLUSIONS: Our study provides evidence for functional HOXB13-MEIS1 interactions in PCa. MEIS1 may contribute to the cancer-promoting actions of HOXB13 in cellular proliferation and gene regulation by prolonging HOXB13 half-life. Our data demonstrates that G84E is not a loss-of-function mutation that interferes with HOXB13 stability or ability to interact with MEIS1.


Subject(s)
Homeodomain Proteins/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression , Gene Expression Profiling , Gene Knockdown Techniques , Germ-Line Mutation , Homeodomain Proteins/genetics , Humans , Male , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , RNA, Messenger/analysis
5.
Gynecol Oncol ; 154(1): 199-206, 2019 07.
Article in English | MEDLINE | ID: mdl-30987772

ABSTRACT

OBJECTIVE: Endocrine therapy is often considered as a treatment for hormone-responsive gynecologic malignancies. In breast cancer, activating mutations in the estrogen receptor (mutESR1) contribute to therapeutic resistance to endocrine therapy, especially aromatase inhibitors (AIs). The purpose of this study was to evaluate the frequency and clinical relevance of ESR1 genomic alterations in gynecologic malignancies. METHODS: DNA from FFPE tumor tissue obtained during routine clinical care for 9645 gynecologic malignancies (ovary, fallopian tube, uterus, cervix, vagina, vulvar, and placenta) was analyzed for all classes of genomic alterations (base substitutions (muts), insertions, deletions, rearrangements, and amplifications) in ESR1 by hybrid capture next generation sequencing. A subset of alterations was characterized in laboratory-based transcription assays for response to endocrine therapies. RESULTS: A total of 295 ESR1 genomic alterations were identified in 285 (3.0%) cases. mutESR1 were present in 86 (0.9%) cases and were more common in uterine compared to other cancers (2.0% vs <1%, respectively p < 0.001). mutESR1 were enriched in carcinomas with endometrioid versus serous histology (4.4% vs 0.2% respectively, p < 0.0001 in uterine and 3.5% vs 0.3% respectively, p = 0.0004 in ovarian carcinomas). In three of four patients with serial sampling, mutESR1 emerged under the selective pressure of AI therapy. Despite decreased potency of estrogen receptor (ER) antagonists in transcriptional assays, clinical benefit was observed following treatment with selective ER-targeted therapy, in one case lasting >48 months. CONCLUSIONS: While the prevalence of ESR1 mutations in gynecologic malignancies is low, there are significant clinical implications useful in guiding therapeutic approaches for these cancers.


Subject(s)
Aromatase Inhibitors/administration & dosage , Estrogen Receptor alpha/genetics , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/genetics , Selective Estrogen Receptor Modulators/administration & dosage , Adult , Aromatase Inhibitors/pharmacology , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm , Female , Humans , Middle Aged , Molecular Targeted Therapy , Mutation , Selective Estrogen Receptor Modulators/pharmacology , Transcription, Genetic/drug effects , Transcriptome , Treatment Outcome , Young Adult
6.
J Biol Chem ; 292(2): 748-759, 2017 Jan 13.
Article in English | MEDLINE | ID: mdl-27903634

ABSTRACT

Targeted inhibitors of the human epidermal growth factor receptor 2 (HER2), such as trastuzumab and lapatinib, are among the first examples of molecularly targeted cancer therapy and have proven largely effective for the treatment of HER2-positive breast cancers. However, approximately half of those patients either do not respond to these therapies or develop secondary resistance. Although a few signaling pathways have been implicated, a comprehensive understanding of mechanisms underlying HER2 inhibitor drug resistance is still lacking. To address this critical question, we undertook a concerted approach using patient expression data sets, HER2-positive cell lines, and tumor samples biopsied both before and after trastuzumab treatment. Together, these methods revealed that high expression and activation of a specific subset of receptor tyrosine kinases (RTKs) was strongly associated with poor clinical prognosis and the development of resistance. Mechanistically, these RTKs are capable of maintaining downstream signal transduction to promote tumor growth via the suppression of cellular senescence. Consequently, these findings provide the rationale for the design of therapeutic strategies for overcoming drug resistance in breast cancer via combinational inhibition of the limited number of targets from this specific subset of RTKs.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Quinazolines/pharmacology , Signal Transduction , Trastuzumab/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Lapatinib , Receptor, ErbB-2 , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Prostate ; 78(4): 266-277, 2018 03.
Article in English | MEDLINE | ID: mdl-29243324

ABSTRACT

BACKGROUND: Whereas the androgen receptor (AR) signaling axis remains a therapeutic target in castration-resistant prostate cancer (CRPC), the emergence of AR mutations and splice variants as mechanisms underlying resistance to contemporary inhibitors of this pathway highlights the need for new therapeutic approaches to target this disease. Of significance in this regard is the considerable preclinical data, indicating that histone deacetylase (HDAC) inhibitors may have utility in the treatment of CRPC. However, the results of clinical studies using HDAC inhibitors (directed against HDAC1, 2, 3, and 8) in CRPC are equivocal, a result that some have attributed to their ability to induce an epithelial to mesenchymal transition (EMT) and neuroendocrine differentiation. We posited that it might be possible to uncouple the beneficial effects of HDAC inhibitors on AR signaling from their undesired activities by targeting specific HDACs as opposed to using the pan-inhibitor strategy that has been employed to date. METHODS: The relative abilities of pan- and selective-Class I HDAC inhibitors to attenuate AR-mediated target gene expression and proliferation were assessed in several prostate cancer cell lines. Small interfering RNA (siRNA)-mediated knockdown approaches were used to confirm the importance of of HDAC 1, 2, and 3 expression in these processes. Further, the ability of each HDAC inhibitor to induce the expression of EMT markers (RNA and protein) and EMT-like phenotype(s) (migration) were also assessed. The anti-tumor efficacy of a HDAC3-selective inhibitor, RGFP966, was compared to the pan-HDAC inhibitor Suberoylanilide Hydroxamic Acid (SAHA) in the 22Rv1 xenograft model. RESULTS: Using genetic and pharmacological approaches we demonstrated that a useful inhibition of AR transcriptional activity, absent the induction of EMT, could be achieved by specifically inhibiting HDAC3. Significantly, we also determined that HDAC3 inhibitors blocked the activity of the constitutively active AR V7-splice variant and inhibited the growth of xenograft tumors expressing this protein. CONCLUSIONS: Our studies provide strong rationale for the near-term development of specific HDAC3 inhibitors for the treatment of CRPC.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Acrylamides/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Cell Migration Assays , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Phenylenediamines/pharmacology , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Androgen/drug effects , Signal Transduction/drug effects , Vorinostat/pharmacology , Xenograft Model Antitumor Assays
8.
Nat Chem Biol ; 12(10): 795-801, 2016 10.
Article in English | MEDLINE | ID: mdl-27501397

ABSTRACT

Clinical resistance to the second-generation antiandrogen enzalutamide in castration-resistant prostate cancer (CRPC), despite persistent androgen receptor (AR) activity in tumors, highlights an unmet medical need for next-generation antagonists. We have identified and characterized tetra-aryl cyclobutanes (CBs) as a new class of competitive AR antagonists that exhibit a unique mechanism of action. These CBs are structurally distinct from current antiandrogens (hydroxyflutamide, bicalutamide, and enzalutamide) and inhibit AR-mediated gene expression, cell proliferation, and tumor growth in several models of CRPC. Conformational profiling revealed that CBs stabilize an AR conformation resembling an unliganded receptor. Using a variety of techniques, it was determined that the AR-CB complex was not recruited to AR-regulated promoters and, like apo AR, remains sequestered in the cytoplasm, bound to heat shock proteins. Thus, we have identified third-generation AR antagonists whose unique mechanism of action suggests that they may have therapeutic potential in CRPC.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology , Structure-Activity Relationship
9.
Mol Cell ; 36(3): 405-16, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19917249

ABSTRACT

HOXB13 is a member of the homeodomain family of sequence-specific transcription factors and, together with the androgen receptor (AR), plays a critical role in the normal development of the prostate gland. We demonstrate here that, in prostate cancer cells, HOXB13 is a key determinant of the response to androgens. Specifically, it was determined that HOXB13 interacts with the DNA-binding domain of AR and inhibits the transcription of genes that contain an androgen-response element (ARE). In contrast, the AR:HOXB13 complex confers androgen responsiveness to promoters that contain a specific HOXB13-response element. Further, HOXB13 and AR synergize to enhance the transcription of genes that contain a HOX element juxtaposed to an ARE. The profound effects of HOXB13 knockdown on androgen-regulated proliferation, migration, and lipogenesis in prostate cancer cells highlight the importance of the observed changes in gene expression.


Subject(s)
Homeodomain Proteins/metabolism , Metribolone/pharmacology , Receptors, Androgen/metabolism , Amino Acid Sequence , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Cluster Analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Humans , Lipid Metabolism/drug effects , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding/drug effects , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
10.
Proc Natl Acad Sci U S A ; 111(9): 3365-70, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550466

ABSTRACT

One of the most widely accepted axioms of mammalian reproductive biology is that pregnancy requires the (sole) support of progesterone, acting in large measure through nuclear progesterone receptors (PRs) in uterine and cervical tissues, without which pregnancy cannot be established or maintained. However, mares lack detectable progesterone in the latter half of pregnancy. Instead of progesterone, several (mainly 5α-reduced) pregnanes are elevated and have long been speculated to provide progestational support in lieu of progesterone itself. To the authors' knowledge, evidence for the bioactivity of a second potent endogenously synthesized pregnane able to support pregnancy in the absence of progesterone has never before been reported. The 5α-reduced progesterone metabolite dihydroprogesterone (DHP) was shown in vivo to stimulate endometrial growth and progesterone-dependent gene expression in the horse at subphysiological concentrations and to maintain equine pregnancy in the absence of luteal progesterone in the third and fourth weeks postbreeding. Results of in vitro studies indicate that DHP is an equally potent and efficacious endogenous progestin in the horse but that the PR evolved with increased agonistic potency for DHP at the expense of potency toward progesterone based on comparisons with human PR responses. Sequence analysis and available literature indicate that the enzyme responsible for DHP synthesis, 5α-reductase type 1, also adapted primarily to metabolize progesterone and thereby to serve diverse roles in the physiology of pregnancy in mammals. Our confirmation that endogenously synthesized DHP is a biopotent progestin in the horse ends decades of speculation, explaining how equine pregnancies survive without measurable circulating progesterone in the last 4 to 5 mo of gestation.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 5-alpha-Dihydroprogesterone/metabolism , Pregnancy/metabolism , Receptors, Progesterone/agonists , 5-alpha-Dihydroprogesterone/blood , Analysis of Variance , Animals , Base Sequence , Chromatography, High Pressure Liquid , Female , Horses , Humans , Immunohistochemistry , Molecular Sequence Data , Progesterone/blood , Progesterone/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Species Specificity , Tandem Mass Spectrometry
11.
J Biol Chem ; 290(29): 17985-17998, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26048986

ABSTRACT

Pregnancy promotes physiological adaptations throughout the body, mediated by the female sex hormones progesterone and estrogen. Changes in the metabolic properties of skeletal muscle enable the female body to cope with the physiological challenges of pregnancy and may also be linked to the development of insulin resistance. We conducted global microarray, proteomic, and metabolic analyses to study the role of the progesterone receptor and its transcriptional regulator, smoothelin-like protein 1 (SMTNL1) in the adaptation of skeletal muscle to pregnancy. We demonstrate that pregnancy promotes fiber-type changes from an oxidative to glycolytic isoform in skeletal muscle. This phenomenon is regulated through an interaction between SMTNL1 and progesterone receptor, which alters the expression of contractile and metabolic proteins. smtnl1(-/-) mice are metabolically less efficient and show impaired glucose tolerance. Pregnancy antagonizes these effects by inducing metabolic activity and increasing glucose tolerance. Our results suggest that SMTNL1 has a role in mediating the actions of steroid hormones to promote fiber switching in skeletal muscle during pregnancy. Our findings also bear on the management of gestational diabetes that develops as a complication of pregnancy in ~4% of women.


Subject(s)
Gene Deletion , Glycolysis , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Phosphoproteins/genetics , Animals , Estrogen Receptor alpha/analysis , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Glucose Tolerance Test , Humans , Insulin Resistance , Mice , Muscle Proteins/metabolism , Muscle, Skeletal/ultrastructure , Oxygen Consumption , Phosphoproteins/metabolism , Pregnancy , Proteomics , Receptors, Progesterone/analysis , Receptors, Progesterone/metabolism
12.
J Pathol ; 235(1): 101-12, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25186463

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a heterodimeric transcriptional regulator with pleiotropic functions in xenobiotic metabolism and detoxification, vascular development and cancer. Herein, we report a previously undescribed role for the AhR signalling pathway in the pathogenesis of the wet, neovascular subtype of age-related macular degeneration (AMD), the leading cause of vision loss in the elderly in the Western world. Comparative analysis of gene expression profiles of aged AhR(-/-) and wild-type (wt) mice, using high-throughput RNA sequencing, revealed differential modulation of genes belonging to several AMD-related pathogenic pathways, including inflammation, angiogenesis and extracellular matrix regulation. To investigate AhR regulation of these pathways in wet AMD, we experimentally induced choroidal neovascular lesions in AhR(-/-) mice and found that they measured significantly larger in area and volume compared to age-matched wt mice. Furthermore, these lesions displayed a higher number of ionized calcium-binding adaptor molecule 1-positive (Iba1(+) ) microglial cells and a greater amount of collagen type IV deposition, events also seen in human wet AMD pathology specimens. Consistent with our in vivo observations, AhR knock-down was sufficient to increase choroidal endothelial cell migration and tube formation in vitro. Moreover, AhR knock-down caused an increase in collagen type IV production and secretion in both retinal pigment epithelial (RPE) and choroidal endothelial cell cultures, increased expression of angiogenic and inflammatory molecules, including vascular endothelial growth factor A (VEGFA) and chemokine (C-C motif) ligand 2 (CCL2) in RPE cells, and increased expression of secreted phosphoprotein 1 (SPP1) and transforming growth factor-ß1 (TGFß1) in choroidal endothelial cells. Collectively, our findings identify AhR as a regulator of multiple pathogenic pathways in experimentally induced choroidal neovascularization, findings that are consistent with a possible role of AhR in wet AMD. The data discussed in this paper have been deposited in NCBI's Gene Expression Omnibus; GEO Submission No. GSE56983, NCBI Tracking System No. 17021116.


Subject(s)
Choroidal Neovascularization/genetics , Gene Expression Regulation/genetics , Receptors, Aryl Hydrocarbon/genetics , Retinal Pigment Epithelium/metabolism , Animals , Cell Movement/genetics , Cells, Cultured , Choroid , Choroidal Neovascularization/metabolism , Humans , Macular Degeneration/genetics , Macular Degeneration/immunology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Aryl Hydrocarbon/metabolism , Retinal Pigment Epithelium/immunology , Vascular Endothelial Growth Factor A/metabolism
13.
Proc Natl Acad Sci U S A ; 110(43): E4069-78, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24106308

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a nuclear receptor that regulates xenobiotic metabolism and detoxification. Herein, we report a previously undescribed role for the AhR signaling pathway as an essential defense mechanism in the pathogenesis of early dry age-related macular degeneration (AMD), the leading cause of vision loss in the elderly. We found that AhR activity and protein levels in human retinal pigment epithelial (RPE) cells, cells vulnerable in AMD, decrease with age. This finding is significant given that age is the most established risk factor for development of AMD. Moreover, AhR(-/-) mice exhibit decreased visual function and develop dry AMD-like pathology, including disrupted RPE cell tight junctions, accumulation of RPE cell lipofuscin, basal laminar and linear-like deposit material, Bruch's membrane thickening, and progressive RPE and choroidal atrophy. High-serum low-density lipoprotein levels were also observed in AhR(-/-) mice. In its oxidized form, this lipoprotein can stimulate increased secretion of extracellular matrix molecules commonly found in deposits from RPE cells, in an AhR-dependent manner. This study demonstrates the importance of cellular clearance via the AhR signaling pathway in dry AMD pathogenesis, implicating AhR as a potential target, and the mouse model as a useful platform for validating future therapies.


Subject(s)
Aging/metabolism , Disease Models, Animal , Macular Degeneration/metabolism , Pigment Epithelium of Eye/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Adolescent , Adult , Aging/genetics , Aging/pathology , Animals , Bruch Membrane/metabolism , Bruch Membrane/pathology , Bruch Membrane/ultrastructure , Cell Line , Child , Extracellular Matrix/metabolism , Female , Humans , Lipofuscin/metabolism , Lipoproteins, LDL/blood , Lipoproteins, LDL/metabolism , Macular Degeneration/genetics , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Middle Aged , Pigment Epithelium of Eye/pathology , Pigment Epithelium of Eye/ultrastructure , RNA Interference , Receptors, Aryl Hydrocarbon/genetics , Tight Junctions/metabolism , Tight Junctions/pathology , Young Adult
14.
Angew Chem Int Ed Engl ; 54(33): 9659-62, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26083457

ABSTRACT

Androgen receptor (AR)-dependent transcription is a major driver of prostate tumor cell proliferation. Consequently, it is the target of several antitumor chemotherapeutic agents, including the AR antagonist MDV3100/enzalutamide. Recent studies have shown that a single AR mutation (F876L) converts MDV3100 action from an antagonist to an agonist. Here we describe the generation of a novel class of selective androgen receptor degraders (SARDs) to address this resistance mechanism. Molecules containing hydrophobic degrons linked to small-molecule AR ligands induce AR degradation, reduce expression of AR target genes and inhibit proliferation in androgen-dependent prostate cancer cell lines. These results suggest that selective AR degradation may be an effective therapeutic prostate tumor strategy in the context of AR mutations that confer resistance to second-generation AR antagonists.


Subject(s)
Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/pharmacology , Proteolysis/drug effects , Receptors, Androgen/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Humans , Hydrophobic and Hydrophilic Interactions , Male , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/chemistry , Phenylthiohydantoin/pharmacology , Point Mutation , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics
15.
Bioorg Med Chem ; 22(2): 917-26, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24360824

ABSTRACT

A series of unsymmetrically substituted biphenyl compounds was designed as alpha helical proteomimetics with the aim of inhibiting the binding of coactivator proteins to the nuclear hormone receptor coactivator binding domain. These compounds were synthesized in good overall yields in seven steps starting from 2-bromoanisole. The final products were evaluated using cotransfection reporter gene assays and mammalian two-hybrid competitive inhibition assays to demonstrate their effectiveness as competitive binding inhibitors. The results from this study indicate that these proteomimetics possess the ability to inhibit coactivator-receptor interactions, but via a mixed mode of inhibition.


Subject(s)
Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Nuclear Receptor Coactivators/antagonists & inhibitors , Protein Structure, Secondary , Binding, Competitive/drug effects , Biomimetic Materials/chemical synthesis , Biphenyl Compounds/chemical synthesis , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Models, Molecular , Molecular Structure , Nuclear Receptor Coactivators/chemistry , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism , Protein Binding/drug effects , Structure-Activity Relationship
16.
Proc Natl Acad Sci U S A ; 108(45): 18348-53, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-22042850

ABSTRACT

Stimulation of resting CD4(+) T lymphocytes leads to rapid proliferation and differentiation into effector (Teff) or inducible regulatory (Treg) subsets with specific functions to promote or suppress immunity. Importantly, Teff and Treg use distinct metabolic programs to support subset specification, survival, and function. Here, we describe that the orphan nuclear receptor estrogen-related receptor-α (ERRα) regulates metabolic pathways critical for Teff. Resting CD4(+) T cells expressed low levels of ERRα protein that increased on activation. ERRα deficiency reduced activated T-cell numbers in vivo and cytokine production in vitro but did not seem to modulate immunity through inhibition of activating signals or viability. Rather, ERRα broadly affected metabolic gene expression and glucose metabolism essential for Teff. In particular, up-regulation of Glut1 protein, glucose uptake, and mitochondrial processes were suppressed in activated ERRα(-/-) T cells and T cells treated with two chemically independent ERRα inhibitors or by shRNAi. Acute ERRα inhibition also blocked T-cell growth and proliferation. This defect appeared as a result of inadequate glucose metabolism, because provision of lipids, but not increased glucose uptake or pyruvate, rescued ATP levels and cell division. Additionally, we have shown that Treg requires lipid oxidation, whereas Teff uses glucose metabolism, and lipid addition selectively restored Treg--but not Teff--generation after acute ERRα inhibition. Furthermore, in vivo inhibition of ERRα reduced T-cell proliferation and Teff generation in both immunization and experimental autoimmune encephalomyelitis models. Thus, ERRα is a selective transcriptional regulator of Teff metabolism that may provide a metabolic means to modulate immunity.


Subject(s)
Cell Differentiation , Lymphocyte Activation , Receptors, Estrogen/physiology , T-Lymphocytes/immunology , Animals , Cell Proliferation , Glucose/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Polymerase Chain Reaction , RNA Interference , Receptors, Estrogen/genetics , ERRalpha Estrogen-Related Receptor
17.
bioRxiv ; 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38260502

ABSTRACT

Protein translation is an energy-intensive ribosome-driven process that is reduced during nutrient scarcity to conserve cellular resources. During prolonged starvation, cells selectively translate specific proteins to enhance their survival (adaptive translation); however, this process is poorly understood. Accordingly, we analyzed protein translation and mRNA transcription by multiple methods in vitro and in vivo to investigate adaptive hepatic translation during starvation. While acute starvation suppressed protein translation in general, proteomic analysis showed that prolonged starvation selectively induced translation of lysosome and autolysosome proteins. Significantly, the expression of the orphan nuclear receptor, estrogen-related receptor alpha (Esrra) increased during prolonged starvation and served as a master regulator of this adaptive translation by transcriptionally stimulating 60S acidic ribosomal protein P1 (Rplp1) gene expression. Overexpression or siRNA knockdown of Esrra expression in vitro or in vivo led to parallel changes in Rplp1 gene expression, lysosome/autophagy protein translation, and autophagy. Remarkably, we have found that Esrra had dual functions by not only regulating transcription but also controling adaptive translation via the Esrra/Rplp1/lysosome/autophagy pathway during prolonged starvation.

18.
Clin Cancer Res ; 30(6): 1152-1159, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38236581

ABSTRACT

PURPOSE: HOXB13 is an androgen receptor (AR) coregulator specifically expressed in cells of prostatic lineage. We sought to associate circulating tumor cell (CTC) HOXB13 expression with outcomes in men with mCRPC treated with abiraterone or enzalutamide. EXPERIMENTAL DESIGN: We conducted a retrospective analysis of the multicenter prospective PROPHECY trial of mCRPC men (NCT02269982, n = 118) treated with abiraterone/enzalutamide. CTC detection and HOXB13 complementary DNA (cDNA) expression was measured using a modified Adnatest, grouping patients into 3 categories: CTC 0 (undetectable); CTC+ HOXB13 CTC low (<4 copies); or CTC+ HOXB13 CTC high. The HOXB13 threshold was determined by maximally selected rank statistics for prognostic associations with overall survival (OS) and progression-free survival (PFS). RESULTS: We included 102 men with sufficient CTC HOXB13 cDNA, identifying 25%, 31%, and 44% of patients who were CTC 0, CTC+ HOXB13 low, and CTC+ HOXB13 high, respectively. Median OS were 25.7, 27.8, and 12.1 months whereas the median PFS were 9.0, 7.7, and 3.8 months, respectively. In subgroup analysis among men with CellSearch CTCs ≥5 copies/mL and adjusting for prior abi/enza treatment and Halabi clinical risk score, the multivariate HR for HOXB13 CTC detection was 2.39 (95% CI, 1.06-5.40) for OS and 2.78 (95% CI, 1.38-5.59) for PFS, respectively. Low HOXB13 CTC detection was associated with lower CTC PSA, PSMA, AR-FL, and AR-V7 detection, and more liver/lung metastases (41% vs. 25%). CONCLUSIONS: Higher CTC HOXB13 expression is associated with AR-dependent biomarkers in CTCs and is adversely prognostic in the context of potent AR inhibition in men with mCRPC.


Subject(s)
Androstenes , Benzamides , Neoplastic Cells, Circulating , Phenylthiohydantoin , Prostatic Neoplasms, Castration-Resistant , Male , Humans , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , RNA , Prospective Studies , Retrospective Studies , DNA, Complementary/therapeutic use , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Nitriles/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/therapeutic use , Homeodomain Proteins/genetics
19.
bioRxiv ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39005294

ABSTRACT

Endocrine therapies targeting the estrogen receptor (ER/ ESR1 ) are the cornerstone to treat ER-positive breast cancers patients, but resistance often limits their effectiveness. Understanding the molecular mechanisms is thus key to optimize the existing drugs and to develop new ER-modulators. Notable progress has been made although the fragmented way data is reported has reduced their potential impact. Here, we introduce EstroGene2.0, an expanded database of its precursor 1.0 version. EstroGene2.0 focusses on response and resistance to endocrine therapies in breast cancer models. Incorporating multi-omic profiling of 361 experiments from 212 studies across 28 cell lines, a user-friendly browser offers comprehensive data visualization and metadata mining capabilities ( https://estrogeneii.web.app/ ). Taking advantage of the harmonized data collection, our follow-up meta-analysis revealed substantial diversity in response to different classes of ER-modulators including SERMs, SERDs, SERCA and LDD/PROTAC. Notably, endocrine resistant models exhibit a spectrum of transcriptomic alterations including a contra-directional shift in ER and interferon signaling, which is recapitulated clinically. Furthermore, dissecting multiple ESR1 -mutant cell models revealed the different clinical relevance of genome-edited versus ectopic overexpression model engineering and identified high-confidence mutant-ER targets, such as NPY1R. These examples demonstrate how EstroGene2.0 helps investigate breast cancer's response to endocrine therapies and explore resistance mechanisms.

20.
Trends Endocrinol Metab ; 34(1): 5-20, 2023 01.
Article in English | MEDLINE | ID: mdl-36443206

ABSTRACT

Gender differences in the functionality of the immune system have been attributed, in part, to direct and indirect effects of sex steroids, especially estrogens, on immune cell repertoire and activity. Notable are studies that have defined roles for estrogens in the regulation of the biology of dendritic cells (DCs), macrophages, T cells and natural killer (NK) cells. Although estrogens can modulate eosinophil function, the mechanisms by which this occurs and how it contributes to the pathobiology of different diseases remains underexplored. Furthermore, although the importance of eosinophils in infection is well established, it remains unclear as to how these innate immune cells, which are present in different tumors, impact the biology of cancer cells and/or response to therapeutics. The observation that eosinophilia influences the efficacy of immune checkpoint blockers (ICBs) is significant considering the role of estrogens as regulators of eosinophil function and recent studies suggesting that response to ICBs is impacted by gender. Thus, in this review, we consider what is known about the roles of estrogen(s) in regulating tissue eosinophilia/eosinophil function and how this influences the pathobiology of breast cancer (in particular). This information provides the context for a discussion of how estrogens/the estrogen receptor (ER) signaling axis can be targeted in eosinophils and how this would be expected to influence the activity of standard-of-care interventions and contemporary immunotherapy regimens in cancer(s).


Subject(s)
Breast Neoplasms , Eosinophilia , Humans , Female , Estrogens/pharmacology , Gonadal Steroid Hormones , Eosinophils/physiology
SELECTION OF CITATIONS
SEARCH DETAIL