Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
bioRxiv ; 2024 May 07.
Article En | MEDLINE | ID: mdl-38766099

Castration resistant prostate cancer (CRPC) remains an incurable disease stage with ineffective treatments options. Here, the androgen receptor (AR) coactivators CBP/p300, which are histone acetyltransferases, were identified as critical mediators of DNA damage repair (DDR) to potentially enhance therapeutic targeting of CRPC. Key findings demonstrate that CBP/p300 expression increases with disease progression and selects for poor prognosis in metastatic disease. CBP/p300 bromodomain inhibition enhances response to standard of care therapeutics. Functional studies, CBP/p300 cistrome mapping, and transcriptome in CRPC revealed that CBP/p300 regulates DDR. Further mechanistic investigation showed that CBP/p300 attenuation via therapeutic targeting and genomic knockdown decreases homologous recombination (HR) factors in vitro, in vivo, and in human prostate cancer (PCa) tumors ex vivo. Similarly, CBP/p300 expression in human prostate tissue correlates with HR factors. Lastly, targeting CBP/p300 impacts HR-mediate repair and patient outcome. Collectively, these studies identify CBP/p300 as drivers of PCa tumorigenesis and lay the groundwork to optimize therapeutic strategies for advanced PCa via CBP/p300 inhibition, potentially in combination with AR-directed and DDR therapies.

2.
Genome Biol ; 25(1): 81, 2024 Mar 29.
Article En | MEDLINE | ID: mdl-38553769

The use of single-cell technologies for clinical applications requires disconnecting sampling from downstream processing steps. Early sample preservation can further increase robustness and reproducibility by avoiding artifacts introduced during specimen handling. We present FixNCut, a methodology for the reversible fixation of tissue followed by dissociation that overcomes current limitations. We applied FixNCut to human and mouse tissues to demonstrate the preservation of RNA integrity, sequencing library complexity, and cellular composition, while diminishing stress-related artifacts. Besides single-cell RNA sequencing, FixNCut is compatible with multiple single-cell and spatial technologies, making it a versatile tool for robust and flexible study designs.


Genomics , RNA , Humans , Animals , Mice , Tissue Fixation/methods , Reproducibility of Results , Sequence Analysis, RNA/methods , RNA/genetics , Genomics/methods , Single-Cell Analysis/methods
3.
Br J Cancer ; 129(8): 1350-1361, 2023 10.
Article En | MEDLINE | ID: mdl-37673961

BACKGROUND: Resistance to androgen receptor signalling inhibitors (ARSIs) represents a major clinical challenge in prostate cancer. We previously demonstrated that the ARSI enzalutamide inhibits only a subset of all AR-regulated genes, and hypothesise that the unaffected gene networks represent potential targets for therapeutic intervention. This study identified the hyaluronan-mediated motility receptor (HMMR) as a survival factor in prostate cancer and investigated its potential as a co-target for overcoming resistance to ARSIs. METHODS: RNA-seq, RT-qPCR and Western Blot were used to evaluate the regulation of HMMR by AR and ARSIs. HMMR inhibition was achieved via siRNA knockdown or pharmacological inhibition using 4-methylumbelliferone (4-MU) in prostate cancer cell lines, a mouse xenograft model and patient-derived explants (PDEs). RESULTS: HMMR was an AR-regulated factor that was unaffected by ARSIs. Genetic (siRNA) or pharmacological (4-MU) inhibition of HMMR significantly suppressed growth and induced apoptosis in hormone-sensitive and enzalutamide-resistant models of prostate cancer. Mechanistically, 4-MU inhibited AR nuclear translocation, AR protein expression and subsequent downstream AR signalling. 4-MU enhanced the growth-suppressive effects of 3 different ARSIs in vitro and, in combination with enzalutamide, restricted proliferation of prostate cancer cells in vivo and in PDEs. CONCLUSION: Co-targeting HMMR and AR represents an effective strategy for improving response to ARSIs.


Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Mice , Animals , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Cell Line, Tumor , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Nitriles/pharmacology , RNA, Small Interfering/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Drug Resistance, Neoplasm/genetics , Cell Proliferation
4.
Elife ; 102021 08 12.
Article En | MEDLINE | ID: mdl-34382934

Alterations to the androgen receptor (AR) signalling axis and cellular metabolism are hallmarks of prostate cancer. This study provides insight into both hallmarks by uncovering a novel link between AR and the pentose phosphate pathway (PPP). Specifically, we identify 6-phosphogluoconate dehydrogenase (6PGD) as an androgen-regulated gene that is upregulated in prostate cancer. AR increased the expression of 6PGD indirectly via activation of sterol regulatory element binding protein 1 (SREBP1). Accordingly, loss of 6PGD, AR or SREBP1 resulted in suppression of PPP activity as revealed by 1,2-13C2 glucose metabolic flux analysis. Knockdown of 6PGD also impaired growth and elicited death of prostate cancer cells, at least in part due to increased oxidative stress. We investigated the therapeutic potential of targeting 6PGD using two specific inhibitors, physcion and S3, and observed substantial anti-cancer activity in multiple models of prostate cancer, including aggressive, therapy-resistant models of castration-resistant disease as well as prospectively collected patient-derived tumour explants. Targeting of 6PGD was associated with two important tumour-suppressive mechanisms: first, increased activity of the AMP-activated protein kinase (AMPK), which repressed anabolic growth-promoting pathways regulated by acetyl-CoA carboxylase 1 (ACC1) and mammalian target of rapamycin complex 1 (mTORC1); and second, enhanced AR ubiquitylation, associated with a reduction in AR protein levels and activity. Supporting the biological relevance of positive feedback between AR and 6PGD, pharmacological co-targeting of both factors was more effective in suppressing the growth of prostate cancer cells than single-agent therapies. Collectively, this work provides new insight into the dysregulated metabolism of prostate cancer and provides impetus for further investigation of co-targeting AR and the PPP as a novel therapeutic strategy.


Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Cell Line , Emodin/analogs & derivatives , Feedback , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Pentose Phosphate Pathway , Prostatic Neoplasms/genetics , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism
5.
Cancer Res ; 81(7): 1704-1718, 2021 04 01.
Article En | MEDLINE | ID: mdl-33547161

The androgen receptor (AR) is the key oncogenic driver of prostate cancer, and despite implementation of novel AR targeting therapies, outcomes for metastatic disease remain dismal. There is an urgent need to better understand androgen-regulated cellular processes to more effectively target the AR dependence of prostate cancer cells through new therapeutic vulnerabilities. Transcriptomic studies have consistently identified lipid metabolism as a hallmark of enhanced AR signaling in prostate cancer, yet the relationship between AR and the lipidome remains undefined. Using mass spectrometry-based lipidomics, this study reveals increased fatty acyl chain length in phospholipids from prostate cancer cells and patient-derived explants as one of the most striking androgen-regulated changes to lipid metabolism. Potent and direct AR-mediated induction of ELOVL fatty acid elongase 5 (ELOVL5), an enzyme that catalyzes fatty acid elongation, was demonstrated in prostate cancer cells, xenografts, and clinical tumors. Assessment of mRNA and protein in large-scale data sets revealed ELOVL5 as the predominant ELOVL expressed and upregulated in prostate cancer compared with nonmalignant prostate. ELOVL5 depletion markedly altered mitochondrial morphology and function, leading to excess generation of reactive oxygen species and resulting in suppression of prostate cancer cell proliferation, 3D growth, and in vivo tumor growth and metastasis. Supplementation with the monounsaturated fatty acid cis-vaccenic acid, a direct product of ELOVL5 elongation, reversed the oxidative stress and associated cell proliferation and migration effects of ELOVL5 knockdown. Collectively, these results identify lipid elongation as a protumorigenic metabolic pathway in prostate cancer that is androgen-regulated, critical for metastasis, and targetable via ELOVL5. SIGNIFICANCE: This study identifies phospholipid elongation as a new metabolic target of androgen action that is critical for prostate tumor metastasis.


Fatty Acid Elongases/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , RNA, Small Interfering/therapeutic use , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Fatty Acid Elongases/genetics , Fatty Acid Elongases/physiology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering/pharmacology , Receptors, Androgen/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Adv Healthc Mater ; 10(6): e2001594, 2021 03.
Article En | MEDLINE | ID: mdl-33274851

Over the last thirty years, research in nanomedicine has widely been focused on applications in cancer therapeutics. However, despite the plethora of reported nanoscale drug delivery systems that can successfully eradicate solid tumor xenografts in vivo, many of these formulations have not yet achieved clinical translation. This issue particularly pertains to the delivery of small interfering RNA (siRNA), a highly attractive tool for selective gene targeting. One of the likely reasons behind the lack of translation is that current in vivo models fail to recapitulate critical elements of clinical solid tumors that may influence drug response, such as cellular heterogeneity in the tumor microenvironment. This study incorporates a more clinically relevant model for assessing siRNA delivery systems; ex vivo culture of prostate cancer harvested from patients who have undergone radical prostatectomy, denoted patient-derived explants (PDE). The model retains native human tissue architecture, microenvironment, and cell signaling pathways. Porous silicon nanoparticles (pSiNPs) behavior in this model is investigated and compared with commonly used 3D cancer cell spheroids for their efficacy in the delivery of siRNA directed against the androgen receptor (AR), a key driver of prostate cancer.


Nanoparticles , Prostatic Neoplasms , Cell Line, Tumor , Humans , Male , Nanomedicine , Prostatic Neoplasms/therapy , RNA, Small Interfering , Tumor Microenvironment
7.
Mol Oncol ; 12(9): 1608-1622, 2018 09.
Article En | MEDLINE | ID: mdl-30117261

Breast and prostate cancer research to date has largely been predicated on the use of cell lines in vitro or in vivo. These limitations have led to the development of more clinically relevant models, such as organoids or murine xenografts that utilize patient-derived material; however, issues related to low take rate, long duration of establishment, and the associated costs constrain use of these models. This study demonstrates that ex vivo culture of freshly resected breast and prostate tumor specimens obtained from surgery, termed patient-derived explants (PDEs), provides a high-throughput and cost-effective model that retains the native tissue architecture, microenvironment, cell viability, and key oncogenic drivers. The PDE model provides a unique approach for direct evaluation of drug responses on an individual patient's tumor, which is amenable to analysis using contemporary genomic technologies. The ability to rapidly evaluate drug efficacy in patient-derived material has high potential to facilitate implementation of personalized medicine approaches.


Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Patient-Specific Modeling , Precision Medicine/methods , Prostatic Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Epithelial Cells , Estrogen Receptor alpha/metabolism , Female , Gelatin Sponge, Absorbable , Heterografts , Humans , Ki-67 Antigen/biosynthesis , Male , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Organoids , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction , Translational Research, Biomedical , Tumor Cells, Cultured , Tumor Microenvironment
8.
Oncotarget ; 6(42): 44728-44, 2015 Dec 29.
Article En | MEDLINE | ID: mdl-26554309

The importance of androgen receptor (AR) signaling is increasingly being recognized in breast cancer, which has elicited clinical trials aimed at assessing the efficacy of androgen deprivation therapy (ADT) for metastatic disease. In prostate cancer, resistance to ADT is frequently associated with the emergence of androgen-independent splice variants of the AR (AR variants, AR-Vs) that lack the LBD and are constitutively active. Women with breast cancer may be prone to a similar phenomenon. Herein, we show that in addition to the prototypical transcript, the AR gene produces a diverse range of AR-V transcripts in primary breast tumors. The most frequently and highly expressed variant was AR-V7 (exons 1/2/3/CE3), which was detectable at the mRNA level in > 50% of all breast cancers and at the protein level in a subset of ERα-negative tumors. Functionally, AR-V7 is a constitutively active and ADT-resistant transcription factor that promotes growth and regulates a transcriptional program distinct from AR in ERα-negative breast cancer cells. Importantly, we provide ex vivo evidence that AR-V7 is upregulated by the AR antagonist enzalutamide in primary breast tumors. These findings have implications for treatment response in the ongoing clinical trials of ADT in breast cancer.


Breast Neoplasms/metabolism , Receptors, Androgen/metabolism , Androgen Antagonists/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Benzamides , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Databases, Genetic , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MCF-7 Cells , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Protein Isoforms , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/drug effects , Receptors, Androgen/genetics , Signal Transduction , Time Factors , Transcription, Genetic , Transfection
9.
Menopause ; 21(1): 79-88, 2014 Jan.
Article En | MEDLINE | ID: mdl-23715406

OBJECTIVE: Medroxyprogesterone acetate (MPA), a component of combined estrogen-progestin therapy (EPT), has been associated with increased breast cancer risk in EPT users. MPA can bind to the androgen receptor (AR), and AR signaling inhibits cell growth in breast tissues. Therefore, the aim of this study was to investigate the potential of MPA to disrupt AR signaling in an ex vivo culture model of normal human breast tissue. METHODS: Histologically normal breast tissues from women undergoing breast surgical operation were cultured in the presence or in the absence of the native AR ligand 5α-dihydrotestosterone (DHT), MPA, or the AR antagonist bicalutamide. Ki67, bromodeoxyuridine, B-cell CLL/lymphoma 2 (BCL2), AR, estrogen receptor α, and progesterone receptor were detected by immunohistochemistry. RESULTS: DHT inhibited the proliferation of breast epithelial cells in an AR-dependent manner within tissues from postmenopausal women, and MPA significantly antagonized this androgenic effect. These hormonal responses were not commonly observed in cultured tissues from premenopausal women. In tissues from postmenopausal women, DHT either induced or repressed BCL2 expression, and the antiandrogenic effect of MPA on BCL2 was variable. MPA significantly opposed the positive effect of DHT on AR stabilization, but these hormones had no significant effect on estrogen receptor α or progesterone receptor levels. CONCLUSIONS: In a subset of postmenopausal women, MPA exerts an antiandrogenic effect on breast epithelial cells that is associated with increased proliferation and destabilization of AR protein. This activity may contribute mechanistically to the increased risk of breast cancer in women taking MPA-containing EPT.


Antineoplastic Agents, Hormonal/pharmacology , Breast/drug effects , Epithelial Cells/drug effects , Medroxyprogesterone Acetate/pharmacology , Androgen Antagonists/pharmacology , Androgens/pharmacology , Anilides/pharmacology , Breast/anatomy & histology , Breast/cytology , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Dihydrotestosterone/pharmacology , Epithelial Cells/physiology , Estrogen Receptor alpha/metabolism , Female , Humans , Ki-67 Antigen/metabolism , Nitriles/pharmacology , Postmenopause , Premenopause/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Androgen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Tissue Culture Techniques , Tosyl Compounds/pharmacology
10.
Prostate ; 73(2): 182-93, 2013 Jan.
Article En | MEDLINE | ID: mdl-22782870

BACKGROUND: Krüppel-like factor (KLF) 6 is a candidate tumor suppressor gene in prostate cancer, but the mechanisms contributing to its loss of expression are poorly understood. We characterized KLF6 expression and DNA methylation status during prostate tumorigenesis in humans and mice. METHODS: KLF6 expression was assessed in matched human non-malignant (NM) and tumor prostate tissues (n = 22) by quantitative real-time PCR (qPCR) and in three independent human prostate cancer cohorts bioinformatically. QPCR for KLF6 expression and methylation-sensitive PCR (MSP) were performed in human prostate LNCaP cancer cells after 5-aza-2'-deoxycytidine treatment. Klf6 protein levels and DNA promoter methylation were assessed in TRansgenic Adenocarcinoma of Mouse Prostate (TRAMP) tumors by immunohistochemistry and MSP, respectively. RESULTS: KLF6 splice variants expression was increased (P = 0.0015) in human prostate tumors compared to NM tissues. Overall, KLF6 was decreased in metastatic compared to primary prostate cancers and reduced expression in primary tumors was associated with a shorter time to relapse (P = 0.0028). Treatment with the demethylating agent 5-aza-2'-deoxycytidine resulted in up-regulation of KLF6 expression (two-fold; P = 0.002) and a decrease in DNA methylation of the KLF6 promoter in LNCaP cells. Klf6 protein levels significantly decreased with progression in the TRAMP model of prostate cancer (P < 0.05), but there was no difference in Klf6 promoter methylation. CONCLUSION: KLF6 expression was decreased in both clinical prostate cancer and the TRAMP model with disease progression, but this could not be explained by DNA methylation of the KLF6 promoter.


Disease Progression , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Animals , Cell Line, Tumor , Cohort Studies , Down-Regulation/genetics , Humans , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/biosynthesis , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
11.
Cancer Epidemiol Biomarkers Prev ; 19(10): 2611-22, 2010 Oct.
Article En | MEDLINE | ID: mdl-20841388

BACKGROUND: Epigenetic alterations are common in prostate cancer, yet how these modifications contribute to carcinogenesis is poorly understood. We investigated whether specific histone modifications are prognostic for prostate cancer relapse, and whether the expression of epigenetic genes is altered in prostate tumorigenesis. METHODS: Global levels of histone H3 lysine-18 acetylation (H3K18Ac) and histone H3 lysine-4 dimethylation (H3K4diMe) were assessed immunohistochemically in a prostate cancer cohort of 279 cases. Epigenetic gene expression was investigated in silico by analysis of microarray data from 23 primary prostate cancers (8 with biochemical recurrence and 15 without) and 7 metastatic lesions. RESULTS: H3K18Ac and H3K4diMe are independent predictors of relapse-free survival, with high global levels associated with a 1.71-fold (P < 0.0001) and 1.80-fold (P = 0.006) increased risk of tumor recurrence, respectively. High levels of both histone modifications were associated with a 3-fold increased risk of relapse (P < 0.0001). Epigenetic gene expression profiling identified a candidate gene signature (DNMT3A, MBD4, MLL2, MLL3, NSD1, and SRCAP), which significantly discriminated nonmalignant from prostate tumor tissue (P = 0.0063) in an independent cohort. CONCLUSIONS: This study has established the importance of histone modifications in predicting prostate cancer relapse and has identified an epigenetic gene signature associated with prostate tumorigenesis. IMPACT: Our findings suggest that targeting the epigenetic enzymes specifically involved in a particular solid tumor may be a more effective approach. Moreover, testing for aberrant expression of epigenetic genes such as those identified in this study may be beneficial in predicting individual patient response to epigenetic therapies.


Histones/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Cohort Studies , Disease Progression , Disease-Free Survival , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Male , Microarray Analysis , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/surgery
12.
Cell Stem Cell ; 6(6): 547-56, 2010 Jun 04.
Article En | MEDLINE | ID: mdl-20569692

Embryonic stem cells (ESCs) are pluripotent, self-renewing, and have the ability to reprogram differentiated cell types to pluripotency upon cellular fusion. Polycomb-group (PcG) proteins are important for restraining the inappropriate expression of lineage-specifying factors in ESCs. To investigate whether PcG proteins are required for establishing, rather than maintaining, the pluripotent state, we compared the ability of wild-type, PRC1-, and PRC2-depleted ESCs to reprogram human lymphocytes. We show that ESCs lacking either PRC1 or PRC2 are unable to successfully reprogram B cells toward pluripotency. This defect is a direct consequence of the lack of PcG activity because it could be efficiently rescued by reconstituting PRC2 activity in PRC2-deficient ESCs. Surprisingly, the failure of PRC2-deficient ESCs to reprogram somatic cells is functionally dominant, demonstrating a critical requirement for PcG proteins in the chromatin-remodeling events required for the direct conversion of differentiated cells toward pluripotency.


B-Lymphocytes/metabolism , Embryonic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , B-Lymphocytes/pathology , Cell Fusion , Cell Line, Transformed , Cellular Reprogramming/genetics , Embryonic Stem Cells/pathology , Gene Knockout Techniques , Histone-Lysine N-Methyltransferase/genetics , Humans , Induced Pluripotent Stem Cells/pathology , Mice , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Repressor Proteins/genetics , Telomerase/biosynthesis , Telomerase/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
13.
Nature ; 460(7253): 410-3, 2009 Jul 16.
Article En | MEDLINE | ID: mdl-19458616

Cohesin-mediated sister chromatid cohesion is essential for chromosome segregation and post-replicative DNA repair. In addition, evidence from model organisms and from human genetics suggests that cohesin is involved in the control of gene expression. This non-canonical role has recently been rationalized by the findings that mammalian cohesin complexes are recruited to a subset of DNase I hypersensitive sites and to conserved noncoding sequences by the DNA-binding protein CTCF. CTCF functions at insulators (which control interactions between enhancers and promoters) and at boundary elements (which demarcate regions of distinct chromatin structure), and cohesin contributes to its enhancer-blocking activity. The underlying mechanisms remain unknown, and the full spectrum of cohesin functions remains to be determined. Here we show that cohesin forms the topological and mechanistic basis for cell-type-specific long-range chromosomal interactions in cis at the developmentally regulated cytokine locus IFNG. Hence, the ability of cohesin to constrain chromosome topology is used not only for the purpose of sister chromatid cohesion, but also to dynamically define the spatial conformation of specific loci. This new aspect of cohesin function is probably important for normal development and disease.


Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/genetics , Chromosomes/metabolism , Gene Expression Regulation, Developmental , Interferon-gamma/genetics , Animals , CCCTC-Binding Factor , CD4-Positive T-Lymphocytes/metabolism , Cell Line , DNA-Binding Proteins , Histones/metabolism , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity , Phosphoproteins/genetics , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Cohesins
14.
PLoS Genet ; 4(9): e1000170, 2008 Sep 05.
Article En | MEDLINE | ID: mdl-18773085

Differentiated cells can be reprogrammed through the formation of heterokaryons and hybrid cells when fused with embryonic stem (ES) cells. Here, we provide evidence that conversion of human B-lymphocytes towards a multipotent state is initiated much more rapidly than previously thought, occurring in transient heterokaryons before nuclear fusion and cell division. Interestingly, reprogramming of human lymphocytes by mouse ES cells elicits the expression of a human ES-specific gene profile, in which markers of human ES cells are expressed (hSSEA4, hFGF receptors and ligands), but markers that are specific to mouse ES cells are not (e.g., Bmp4 and LIF receptor). Using genetically engineered mouse ES cells, we demonstrate that successful reprogramming of human lymphocytes is independent of Sox2, a factor thought to be required for induced pluripotent stem (iPS) cells. In contrast, there is a distinct requirement for Oct4 in the establishment but not the maintenance of the reprogrammed state. Experimental heterokaryons, therefore, offer a powerful approach to trace the contribution of individual factors to the reprogramming of human somatic cells towards a multipotent state.


B-Lymphocytes/cytology , Cellular Reprogramming/physiology , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , HMGB Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Fusion , Cell Nucleus/metabolism , Embryonic Stem Cells/cytology , Homeodomain Proteins/metabolism , Humans , Hybrid Cells/metabolism , Mice , Nanog Homeobox Protein , Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors
15.
Biol Reprod ; 74(1): 153-60, 2006 Jan.
Article En | MEDLINE | ID: mdl-16192401

Peroxisome proliferator-activated receptor-gamma (PPARG) and PPAR-alpha (PPARA) control metabolic processes in many cell types and act as anti-inflammatory regulators in macrophages. PPAR-activating ligands include thiazolidinediones (TZDs), such as troglitazone, once frequently used to treat insulin resistance as well as symptoms of polycystic ovary syndrome (PCOS). Since macrophages within the ovary mediate optimal follicle development, TZD actions to improve PCOS symptoms are likely to be partly mediated through these specifically localized immune cells. In mouse ovary, PPARG protein was expressed in granulosa cells and in isolated cells localized to theca, stroma, and corpora lutea, consistent with EMR1+ macrophages. Isolation of immune cells (EMR1+ or H2+) showed that Pparg and Ppara were expressed in ovarian macrophages at much higher levels than in peritoneal macrophages. Ovulatory human chorionic gonadotropin downregulated expression of Pparg and Ppara in EMR1+ ovarian macrophages, but no hormonal responsiveness was observed in H2+ cells. Downstream anti-inflammatory effects of PPARG activation were analyzed by in vitro treatment of isolated macrophages with troglitazone. Interleukin-1 beta (Il1b) expression was not altered, and tumor necrosis factor-alpha (Tnf) expression was affected in peritoneal macrophages only. In ovarian macrophages, inducible nitric oxide synthase (Nos2), an important proinflammatory enzyme that regulates ovulation, was significantly reduced by troglitazone treatment, an effect that was restricted to cells from the preovulatory ovary. Thus, expression of PPARs within ovarian macrophages is hormonally regulated, reflecting the changing roles of these cells during the ovulatory cycle. Additionally, ovarian macrophages respond directly to troglitazone to downregulate expression of proinflammatory Nos2, providing mechanistic information about ovarian effects of TZD treatment.


Chromans/pharmacology , Macrophages/drug effects , Nitric Oxide Synthase Type II/drug effects , Ovary/drug effects , PPAR gamma/drug effects , Thiazolidinediones/pharmacology , Animals , Blotting, Western , Calcium-Binding Proteins , Female , Immunohistochemistry , In Vitro Techniques , Interleukin-1/metabolism , Macrophages/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Ovary/metabolism , Ovulation/metabolism , PPAR alpha/drug effects , PPAR alpha/metabolism , PPAR gamma/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Reverse Transcriptase Polymerase Chain Reaction , Troglitazone , Tumor Necrosis Factor-alpha/metabolism
16.
Hum Reprod Update ; 10(2): 119-33, 2004.
Article En | MEDLINE | ID: mdl-15073142

Macrophages are multifunctional cells that play key roles in the immune response and are abundant throughout female reproductive tissues. Macrophages are identified in tissues by their expression of cell surface receptors and can execute diverse functional activities, including phagocytosis and degradation of foreign antigens, matrix dissolution and tissue remodelling, and production and secretion of cytokines, chemokines and growth factors. Their specific localization and variations in distribution in the ovary during different stages of the cycle, as well as their presence in peri-ovulatory human follicular fluid, suggest that macrophages play diverse roles in intra-ovarian events including folliculogenesis, tissue restructuring at ovulation and corpus luteum formation and regression. This review presents the existing evidence for the regulation of ovarian function by macrophages and macrophage-derived products, highlighting the implications of these cells in ovarian diseases, particularly polycystic ovary syndrome, endometriosis and premature ovarian failure.


Macrophages/physiology , Ovary/physiology , Animals , Antigens/metabolism , Chemokines/metabolism , Cytokines/metabolism , Endometriosis/pathology , Female , Follicular Atresia/physiology , Follicular Fluid/cytology , Growth Substances/metabolism , Humans , Ovulation , Phagocytosis/physiology , Polycystic Ovary Syndrome/pathology , Primary Ovarian Insufficiency/pathology
17.
Endocrinology ; 144(11): 5006-13, 2003 Nov.
Article En | MEDLINE | ID: mdl-12959975

Leptin is an important satiety hormone and reproductive regulator and is found, along with its receptors, throughout the ovary. To date, the changes in ovarian expression of both of these proteins throughout the estrous cycle has not been studied, and the examination of protein expression has not distinguished between different forms of the receptor. In this study leptin mRNA expression in the immature gonadotropin-primed rat ovary increased 3-fold after human chorionic gonadotropin administration, followed by a dramatic increase in mRNA for both the short form (Ob-Ra) and the long form (Ob-Rb) of the leptin receptor (approximately 8- and 7-fold, respectively). A corresponding increase in mRNA expression of the receptor was not observed in isolated preovulatory follicles. Using immunohistochemistry, we observed protein expression of the long form of the leptin receptor (Ob-Rb) in the ovary, with high intensities observed in oocytes and endothelial cells as well as thecal cells and corpora lutea. These results suggest that ovarian expression of leptin and its receptor is regulated across the cycle by gonadotropins, with peak expression at ovulation, indicating a possible involvement in oocyte maturation, angiogenesis, follicle rupture, or subsequent corpus luteum formation.


Leptin/metabolism , Ovary/metabolism , Receptors, Cell Surface/metabolism , Animals , Chorionic Gonadotropin/pharmacology , Culture Techniques , Estradiol/blood , Female , Follicular Phase , Leptin/blood , Leptin/genetics , Organ Size , Ovarian Follicle/metabolism , Ovary/anatomy & histology , Ovary/drug effects , Progesterone/blood , RNA/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Receptors, Leptin , Time Factors
18.
Biol Reprod ; 66(5): 1548-54, 2002 May.
Article En | MEDLINE | ID: mdl-11967222

Leptin is a product of the ob gene that is produced primarily by adipose tissue. Leptin and its receptors are found within the ovary, but it is unclear what function this hormone has in the ovary. Using immunohistochemistry, we determined that leptin is found in most cell types in the murine ovary, with the highest staining levels observed in the oocyte. Leptin receptor was also expressed in all of the main ovarian cell types, with the thecal cell layer exhibiting the highest staining levels. Leptin administration did not affect spontaneous or induced maturation of either isolated denuded oocytes or cumulus-oocyte complexes, but it did significantly increase the rate of meiotic resumption in preovulatory follicle-enclosed oocytes (P < 0.01). Measurements of cAMP within oocytes cultured with leptin showed that this enhanced ability to resume meiosis does not occur via activation of phosphodiesterase 3B and subsequent cAMP reduction. These results provide evidence that leptin affects oocyte maturation when the oocyte is cultured within its normal follicular environment. It is suggested that leptin may induce the production of another factor, possibly from thecal cells, that directly or indirectly acts on the oocyte to initiate germinal vesicle breakdown in this species.


Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Leptin/biosynthesis , Leptin/physiology , Oocytes/growth & development , Ovary/metabolism , Ovary/physiology , Receptors, Cell Surface , Animals , Cells, Cultured , Cyclic AMP/biosynthesis , Female , Immunohistochemistry , Leptin/pharmacology , Mice , Oocytes/metabolism , Ovarian Follicle/physiology , Pregnancy , Receptors, Leptin , Steroids/biosynthesis , Theca Cells/metabolism
...