Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Irina U. Agoulnik is active.

Publication


Featured researches published by Irina U. Agoulnik.


Nature Genetics | 2000

A transgenic insertion upstream of Sox9 is associated with dominant XX sex reversal in the mouse

Colin E. Bishop; Deanne J. Whitworth; Yanjun Qin; Alexander I. Agoulnik; Irina U. Agoulnik; Wilbur R. Harrison; Richard R. Behringer; Paul A. Overbeek

In most mammals, male development is triggered by the transient expression of the Y-chromosome gene, Sry, which initiates a cascade of gene interactions ultimately leading to the formation of a testis from the indifferent fetal gonad. Several genes, in particular Sox9, have a crucial role in this pathway. Despite this, the direct downstream targets of Sry and the nature of the pathway itself remain to be clearly established. We report here a new dominant insertional mutation, Odsex (Ods), in which XX mice carrying a 150-kb deletion (approximately 1 Mb upstream of Sox9) develop as sterile XX males lacking Sry. During embryogenesis, wild-type XX fetal gonads downregulate Sox9 expression, whereas XY and XX Ods/+ fetal gonads upregulate and maintain its expression. We propose that Ods has removed a long-range, gonad-specific regulatory element that mediates the repression of Sox9 expression in XX fetal gonads. This repression would normally be antagonized by Sry protein in XY embryos. Our data are consistent with Sox9 being a direct downstream target of Sry and provide genetic evidence to support a general repressor model of sex determination in mammals.


Cancer Research | 2005

Role of SRC-1 in the Promotion of Prostate Cancer Cell Growth and Tumor Progression

Irina U. Agoulnik; Ajula Vaid; William E. Bingman; Halime Erdeme; Anna Frolov; Carolyn L. Smith; Gustavo Ayala; Michael Ittmann; Nancy L. Weigel

Prostate cancer is initially androgen dependent and there is evidence that androgen receptor continues to play a role in androgen-independent prostate cancer. Androgen receptor activity depends both on the level of androgens and on the level of coactivators that interact with androgen receptor. Our goal was to evaluate the role of the androgen receptor coactivator SRC-1 in prostate cancer progression. Using tissue arrays to measure SRC-1 protein levels, we found that increased SRC-1 expression in clinically localized, androgen-dependent cancer is associated with clinical and pathologic variables of increased tumor aggressiveness. Interestingly, there was variable expression of SRC-1 in normal prostate tissue which correlated with the staining intensity of the corresponding cancer tissue. To test the contribution of SRC-1, we examined its role in androgen-dependent LNCaP and androgen-independent C4-2 prostate cancer cell lines. Using small interfering RNA to reduce expression of androgen receptor, we found that androgen receptor was required both for cell growth and for basal expression of prostate-specific antigen in the androgen-independent C4-2 cell line. Thus, although the cells can grow in an androgen-depleted medium, they remained androgen receptor dependent. Reduction of SRC-1 expression significantly reduced growth and altered androgen receptor target gene regulation in both LNCaP and C4-2 cell lines whereas it had no effect on the growth of the androgen receptor-negative PC-3 and DU145 prostate cancer cell lines. Although the requirement for androgens and androgen receptor in the development of prostate cancer is well established, our study implicates enhanced androgen receptor activity through elevated expression of SRC-1 in the development of more aggressive disease in men with prostate cancer.


Journal of Cellular Biochemistry | 2006

Androgen receptor action in hormone-dependent and recurrent prostate cancer.

Irina U. Agoulnik; Nancy L. Weigel

The importance of androgens and androgen receptors (AR) in primary prostate cancer is well established. Metastatic disease is usually treated with some form of androgen ablation, which is effective for a limited amount of time. The role of AR in prostate cancers that recur despite androgen ablation therapy is less certain. Most of these tumors express prostate specific antigen (PSA), an androgen‐regulated gene; moreover, AR is generally highly expressed in recurrent prostate cancer. We propose that AR continues to play a role in many of these tumors and that it is not only the levels of AR, ligands, and co‐regulators, but also the changes in cell signaling that induce AR action in recurrent prostate cancer. These pathways are, therefore, potential therapeutic targets. J. Cell. Biochem. 99: 362–372, 2006.


Cancer Research | 2011

Decreased Expression and Androgen Regulation of the Tumor Suppressor Gene INPP4B in Prostate Cancer

Myles C. Hodgson; Long Jiang Shao; Anna Frolov; Rile Li; Leif E. Peterson; Gustavo Ayala; Michael Ittmann; Nancy L. Weigel; Irina U. Agoulnik

Patients with metastatic prostate cancer who undergo androgen-ablation therapy invariably relapse and develop incurable castration-resistant disease. Activation of the prosurvival Akt pathway accompanies androgen ablation. We discovered that the androgen receptor induces the expression of the tumor suppressor inositol polyphosphate 4-phosphatase type II (INPP4B) but not PTEN in prostate cancer cells. Optimal induction of INPP4B by an androgen receptor required the expression of the transcriptional coactivator NCoR. INPP4B dephosphorylates phosphatidylinositol-3, 4-bisphosphate, which leads to reduced phosphorylation and activity of Akt. In support of a key role for INPP4B in Akt control, INPP4B depletion activated Akt and increased cellular proliferation. The clinical significance of INPP4B in androgen-dependent prostate cancers was determined in normal or primary tumor prostate tissues derived from radical prostatectomy specimens. In primary tumors, the expression of both INPP4B and PTEN was substantially reduced compared with normal tissue. Further, the decreased expression of INPP4B reduced the time to biochemical recurrence. Thus, androgen ablation can activate the Akt pathway via INPP4B downregulation, thereby mitigating the antitumor effects of androgen ablation. Our findings reinforce the concept that patients undergoing androgen ablation may benefit from Akt-targeting therapies.


Journal of Cellular Biochemistry | 2006

Quantifying effects of ligands on androgen receptor nuclear translocation, intranuclear dynamics, and solubility.

Marco Marcelli; David L. Stenoien; Adam T. Szafran; Silvia Simeoni; Irina U. Agoulnik; Nancy L. Weigel; Tim Moran; Ivana Mikic; Jeffrey H. Price; Michael A. Mancini

Using manual and automated high throughput microscopy (HTM), ligand‐dependent trafficking of green fluorescent protein‐androgen receptor (GFP‐AR) was analyzed in fixed and living cells to determine its spatial distribution, solubility, mobility, and co‐activator interactions. Within minutes, addition of the agonist R1881 resulted translocation of GFP‐AR from the cytoplasm to the nucleus, where it displayed a hyperspeckled pattern and extraction resistance in low expressing cells. AR antagonists (Casodex, hydroxyflutamide) also caused nuclear translocation, however, the antagonist‐bound GFP‐AR had a more diffuse nuclear distribution, distinct from the agonist‐bound GFP‐AR, and was completely soluble; overexpressed GFP‐AR in treated cells was extraction resistant, independent of ligand type. To more dramatically show the different effects of ligand on AR distribution, we utilized an AR with a mutation in the DNA binding domain (ARC619Y) that forms distinct foci upon exposure to agonists but retains a diffuse nuclear distribution in the presence of antagonists. Live‐cell imaging of this mutant demonstrated that cytoplasmic foci formation occurs immediately upon agonist but not antagonist addition. Fluorescence recovery after photobleaching (FRAP) revealed that agonist‐bound GFP‐AR exhibited reduced mobility relative to unliganded or antagonist‐bound GFP‐AR. Importantly, agonist‐bound GFP‐AR mobility was strongly affected by protein expression levels in transiently transfected cells, and displayed reduced mobility even in slightly overexpressing cells. Cyan fluorescent protein‐AR (CFP‐AR) and yellow fluorescent protein‐CREB binding protein (YFP‐CBP) in the presence of agonists and antagonists were used to demonstrate that CFP‐AR specifically co‐localizes with YFP‐CBP in an agonist dependent manner. Dual FRAP experiments demonstrated that CBP mobility mirrored AR mobility only in the presence of agonist. HTM enabled simultaneous studies of the sub‐cellular distribution of GFP‐AR and ARC619Y in response to a range of concentrations of agonists and antagonists (ranging from 10−12 to 10−5) in thousands of cells. These results further support the notion that ligand specific interactions rapidly affect receptor and co‐factor organization, solubility, and molecular dynamics, and each can be aberrantly affected by mutation and overexpression. J. Cell. Biochem. 98: 770–788, 2006.


Cancer Biology & Therapy | 2006

The role of relaxin in endometrial cancer

Aparna A. Kamat; Shu Feng; Irina U. Agoulnik; Farrah Kheradmand; Natalia V. Bogatcheva; Donna Coffey; Anil K. Sood; Alexander I. Agoulnik

Relaxin (RLN) is a naturally occurring hormone that is known to modulate connective tissue remodeling in the uterus and cervix. Our goal was to investigate the role of RLN in endometrial cancer. RLN expression was evaluated using immunohistochemistry in 57 samples of invasive endometrial carcinoma (EC) and 10 benign endometria. 67% of high-stage (III/IV) tumors demonstrated strong RLN expression compared to 37% of low-stage (I/II) cases. Strong RLN expression associated significantly with high-grade and depth of myometrial invasion. Notably, strong RLN expression was associated with a significantly shorter overall survival (p


Endocrine-related Cancer | 2010

Suppression of relaxin receptor RXFP1 decreases prostate cancer growth and metastasis

Shu Feng; Irina U. Agoulnik; Anne Truong; Zhen Li; Chad J. Creighton; Elena M. Kaftanovskaya; Rhea Pereira; Hee Dong Han; Gabriel Lopez-Berestein; Thomas Klonisch; Michael Ittmann; Anil K. Sood; Alexander I. Agoulnik

Relaxin (RLN) is a small peptide hormone expressed in several cancers of reproductive and endocrine organs. Increased expression of RLN in prostate cancer correlates with aggressive cancer. RLN G-protein-coupled receptor (RLN family peptide receptor 1, RXFP1) is expressed in both androgen receptor (AR)-positive and -negative prostate cancers as well as in prostate cancer cell lines. RLN behaves as a cell growth factor and increases invasiveness and proliferation of cancer cells in vitro and in vivo. The objective of this study is to determine whether downregulation of RXFP1 expression using small interfering RNA (siRNA) reduces cancer growth and metastasis in a xenograft model of prostate cancer. We used two well-characterized prostate adenocarcinoma cell lines, AR-positive LNCaP cells and AR-negative PC3 cells. The tumors were established in nude male mice by s.c. injections. Intratumoral injections of siRNAs loaded on biodegradable chitosan nanoparticles led to a downregulation of RXFP1 receptor expression and a dramatic reduction in tumor growth. In LNCaP tumors, the siRNA treatment led to an extensive necrosis. In PC3 xenografts treated with siRNA against RXFP1, the smaller tumor size was associated with the decreased cell proliferation and increased apoptosis. The downregulation of RXFP1 resulted in significant decrease in metastasis rate in PC3 tumors. Global transcriptional profiling of PC3 cells treated with RXFP1 siRNA revealed genes with significantly altered expression profiles previously shown to promote tumorigenesis, including the downregulation of MCAM, MUC1, ANGPTL4, GPI, and TSPAN8. Thus, the suppression of RLN/RXFP1 may have potential therapeutic benefits in prostate cancer.


Steroids | 2009

Coactivator selective regulation of androgen receptor activity

Irina U. Agoulnik; Nancy L. Weigel

The androgen receptor (AR) is a ligand activated nuclear receptor, which regulates transcription and stimulates growth of androgen dependent prostate cancer. To regulate transcription, AR recruits a series of coactivators that modify chromatin and facilitate transcription. However, information on ligand and target gene-specific requirements for coactivators is limited. We compared the actions of the p160 coactivators SRC-1 and SRC-3/RAC3 with SRA (steroid receptor RNA activator). All three coactivate AR in the presence of agonist as expected. However, overexpression of either SRC-1 or SRC-3 increased AR activity in response to the partial antagonist, cyproterone acetate, whereas SRA was unable to stimulate AR activity under these conditions. Using siRNA to reduce expression of these coactivators in LNCaP cells, we also found promoter specific requirement for these coactivators. SRC-3 is required for optimal androgen dependent induction of PSA, TMPRSS2, and PMEPA1 whereas SRA is required only for optimal induction of the TMPRSS2 gene. These data indicate that different groups of AR target genes have distinct requirements for coactivators and response to AR ligands.


Molecular Endocrinology | 2008

Target Gene-Specific Regulation of Androgen Receptor Activity by p42/p44 Mitogen-Activated Protein Kinase

Irina U. Agoulnik; William E. Bingman; Manjula Nakka; Wei Li; Qianben Wang; X. Shirley Liu; Myles Brown; Nancy L. Weigel

Evidence that the androgen receptor (AR) is not only important in androgen-dependent prostate cancer, but also continues to play a role in tumors that become resistant to androgen deprivation therapies, highlights the need to find alternate means to block AR activity. AR, a hormone-activated transcription factor, and its coactivators are phosphoproteins. Thus, we sought to determine whether inhibition of specific cell signaling pathways would reduce AR function. We found that short-term inhibition of p42/p44 MAPK activity either by a MAPK kinase inhibitor, U0126, or by depletion of kinase with small interfering RNA caused target gene-specific reductions in AR activity. AR enhances histone H3 acetylation of target genes that are sensitive to U0126 including prostate-specific antigen and TMPRSS2, but does not increase histone H3 acetylation of the U0126-resistant PMEPA1 gene. Thus, although AR induces transcription of many target genes, the molecular changes induced by AR at the chromatin level are target gene specific. Long-term treatment (24-48 h) with U0126 causes a G1 cell cycle arrest and reduces AR expression both through a decrease in AR mRNA and a reduction in AR protein stability. Thus, treatments that reduce p42/p44 MAPK activity in prostate cancer have the potential to reduce AR activity through a reduction in expression levels as well as by target gene-selective inhibition of AR function.


Biology of Reproduction | 2007

Developmental Expression and Gene Regulation of Insulin-like 3 Receptor RXFP2 in Mouse Male Reproductive Organs

Shu Feng; Natalia V. Bogatcheva; Anne Truong; Borys Korchin; Colin E. Bishop; Thomas Klonisch; Irina U. Agoulnik; Alexander I. Agoulnik

Abstract The mutations of testicular insulin-like 3 (INSL3) hormone or its receptor RXFP2 cause cryptorchidism in male mice. Here we have examined Rxfp2 gene expression at different stages of embryonic and postnatal mouse development in male reproductive tissues employing quantitative RT-PCR and several RXFP2-specific antibodies directed toward different parts of the RXFP2 protein. Receptor expression was markedly increased after birth and was readily detectable in the epididymis, Leydig cells, and germ cells of the testis. The strongest expression was detected in adult mouse cremaster muscle. INSL3 treatment increased cell proliferation of embryonic gubernacular and TM3 embryonic Leydig cells, implicating active INSL3-mediated autocrine signaling in these cells and identifying TM3 as a novel in vitro model to study the effects of RXFP2 signaling. We generated Tg(Rxfp2-cre)Aia (Rxfp2-iCre) transgenic mice expressing improved Cre recombinase (iCre) under the control of the 2.4-kb mouse Rxfp2 promoter. The iCre was expressed in the gubernacular ligament at E14.5, indicating that this promoter is able to drive Rxfp2 gene expression during transabdominal testis descent. We demonstrated that the transcription factor Sox9, a known male sex determination factor, is expressed in mouse embryonic gubernacula and upregulated human, but not mouse, promoter luciferase reporter constructs. In conclusion, we have determined the developmental expression profile of INSL3 receptor employing newly characterized RXFP2 antisera and a novel Rxfp2-iCre transgenic mouse model. We determined the promoter region capable of providing the gubernacular-specific expression of Rxfp2. Analysis of RXFP2 promoter identified SOX9 as a new transcriptional enhancer of human gene expression.

Collaboration


Dive into the Irina U. Agoulnik's collaboration.

Top Co-Authors

Avatar

Alexander I. Agoulnik

Florida International University

View shared research outputs
Top Co-Authors

Avatar

Nancy L. Weigel

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Juan J. Marugan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xin Hu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael Ittmann

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jingbo Xiao

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marc Ferrer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Noel Southall

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shu Feng

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Anna Frolov

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge