Network


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

Hotspot


Dive into the research topics where Emelyn H. Shroff is active.

Publication


Featured researches published by Emelyn H. Shroff.


Journal of Experimental Medicine | 2004

Nuclear Factor of Activated T Cells Balances Angiogenesis Activation and Inhibition

Tetiana Zaichuk; Emelyn H. Shroff; Rebekah Emmanuel; Stephanie Filleur; Thomas Nelius; Olga V. Volpert

It has been demonstrated that vascular endothelial cell growth factor (VEGF) induction of angiogenesis requires activation of the nuclear factor of activated T cells (NFAT). We show that NFATc2 is also activated by basic fibroblast growth factor and blocked by the inhibitor of angiogenesis pigment epithelial–derived factor (PEDF). This suggests a pivotal role for this transcription factor as a convergence point between stimulatory and inhibitory signals in the regulation of angiogenesis. We identified c-Jun NH2-terminal kinases (JNKs) as essential upstream regulators of NFAT activity in angiogenesis. We distinguished JNK-2 as responsible for NFATc2 cytoplasmic retention by PEDF and JNK-1 and JNK-2 as mediators of PEDF-driven NFAT nuclear export. We identified a novel NFAT target, caspase-8 inhibitor cellular Fas-associated death domain–like interleukin 1β–converting enzyme inhibitory protein (c-FLIP), whose expression was coregulated by VEGF and PEDF. Chromatin immunoprecipitation showed VEGF-dependent increase of NFATc2 binding to the c-FLIP promoter in vivo, which was attenuated by PEDF. We propose that one possible mechanism of concerted angiogenesis regulation by activators and inhibitors may be modulation of the endothelial cell apoptosis via c-FLIP controlled by NFAT and its upstream regulator JNK.


PLOS ONE | 2009

Nitric oxide induces cell death by regulating anti-apoptotic BCL-2 family members.

Colleen Snyder; Emelyn H. Shroff; Jing Liu; Navdeep S. Chandel

Nitric oxide (NO) activates the intrinsic apoptotic pathway to induce cell death. However, the mechanism by which this pathway is activated in cells exposed to NO is not known. Here we report that BAX and BAK are activated by NO and that cytochrome c is released from the mitochondria. Cells deficient in Bax and Bak or Caspase-9 are completely protected from NO-induced cell death. The individual loss of the BH3-only proteins, Bim, Bid, Puma, Bad or Noxa, or Bid knockdown in Bim−/−/Puma−/− MEFs, does not prevent NO-induced cell death. Our data show that the anti-apoptotic protein MCL-1 undergoes ASK1-JNK1 mediated degradation upon exposure to NO, and that cells deficient in either Ask1 or Jnk1 are protected against NO-induced cell death. NO can inhibit the mitochondrial electron transport chain resulting in an increase in superoxide generation and peroxynitrite formation. However, scavengers of ROS or peroxynitrite do not prevent NO-induced cell death. Collectively, these data indicate that NO degrades MCL-1 through the ASK1-JNK1 axis to induce BAX/BAK-dependent cell death.


International Journal of Cancer | 2007

Androgen receptor targets NFκB and TSP1 to suppress prostate tumor growth in vivo

Thomas Nelius; Stephanie Filleur; Alexander Yemelyanov; Irina Budunova; Emelyn H. Shroff; Yelena Mirochnik; Arin B. Aurora; Dorina Veliceasa; Wuhan Xiao; Zhou Wang; Olga V. Volpert

The androgen role in the maintenance of prostate epithelium is subject to conflicting opinions. While androgen ablation drives the regression of normal and cancerous prostate, testosterone may cause both proliferation and apoptosis. Several investigators note decreased proliferation and stronger response to chemotherapy of the prostate cancer cells stably expressing androgen receptor (AR), however no mechanistic explanation was offered. In this paper we demonstrate in vivo anti‐tumor effect of the AR on prostate cancer growth and identify its molecular mediators. We analyzed the effect of AR on the tumorigenicity of prostate cancer cells. Unexpectedly, the AR‐expressing cells formed tumors in male mice at a much lower rate than the AR‐negative controls. Moreover, the AR‐expressing tumors showed decreased vascularity and massive apoptosis. AR expression lowered the angiogenic potential of cancer cells, by increasing secretion of an anti‐angiogenic protein, thrombospondin‐1. AR activation caused a decrease in RelA, a subunit of the pro‐survival transcription factor NFκB, reduced its nuclear localization and transcriptional activity. This, in turn, diminished the expression of its anti‐apoptotic targets, Bcl‐2 and IL‐6. Increased apoptosis within AR‐expressing tumors was likely due to the NFκB suppression, since it was restricted to the cells lacking nuclear (active) NFκB. Thus we for the first time identified combined decrease of NFκB and increased TSP1 as molecular events underlying the AR anti‐tumor activity in vivo. Our data indicate that intermittent androgen ablation is preferable to continuous withdrawal, a standard treatment for early‐stage prostate cancer.


The FASEB Journal | 2009

Hyperoxia-induced premature senescence requires p53 and pRb, but not mitochondrial matrix ROS.

Tatyana A. Klimova; Eric L. Bell; Emelyn H. Shroff; Frank Weinberg; Colleen Snyder; Goberdan P. Dimri; Paul T. Schumacker; G. R. Scott Budinger; Navdeep S. Chandel

Senescence is a potential tumor‐suppressing mechanism and a commonly used model of cellular aging. One current hypothesis to explain senescence, based in part on the correlation of oxygen with senescence, postulates that it is caused by oxidative damage from reactive oxygen species (ROS). Here, we further test this theory by determining the mechanisms of hyperoxia‐induced senescence. Exposure to 70% O2 led to stress‐induced, telomere‐independent senescence. Although hyperoxia elevated mitochondrial ROS production, overexpression of antioxidant proteins was not sufficient to prevent hyperoxia‐induced senescence. Hyperoxia activated AMPK however, overexpression of a kinase‐dead mutant of LKB1, which prevented AMPK activation, did not prevent hyperoxia‐induced senescence. Knocking down p21 via shRNA, or suppression of the p16/pRb pathway by either BMI1 or HPV16‐E7 overexpression, was also insufficient to prevent hyperoxia‐induced senescence. However, suppressing p53 function resulted in partial rescue from senescence, suggesting that hyperoxia‐induced senescence involves p53. Suppressing both the p53 and pRb pathways resulted in almost complete protection, indicating that both pathways cooperate in hyperoxia‐induced senescence. Collectively, these results indicate a ROS‐independent but p53/pRb‐dependent senescence mechanism during hyperoxia.— Klimova, T. A., Bell, E. L., Shroff, E. H., Weinberg, F. D., Snyder, C. M., Dimri, G. P., Schumacker, P. T., Budinger, G. R. S., Chandel, N. S. Hyperoxia‐induced premature senescence requires p53 and pRb, but not mitochondrial matrix ROS. FASEB J. 23, 783–794 (2009)


Molecular and Cellular Biology | 2007

Loss of Mcl-1 protein and inhibition of electron transport chain together induce anoxic cell death

Joslyn K. Brunelle; Emelyn H. Shroff; Harris Perlman; Andreas Strasser; Carlos T. Moraes; Richard A. Flavell; Nika N. Danial; Brian Keith; Craig B. Thompson; Navdeep S. Chandel

ABSTRACT How cells die in the absence of oxygen (anoxia) is not understood. Here we report that cells deficient in Bax and Bak or caspase-9 do not undergo anoxia-induced cell death. However, the caspase-9 null cells do not survive reoxygenation due to the generation of mitochondrial reactive oxygen species. The individual loss of Bim, Bid, Puma, Noxa, Bad, caspase-2, or hypoxia-inducible factor 1β, which are potential upstream regulators of Bax or Bak, did not prevent anoxia-induced cell death. Anoxia triggered the loss of the Mcl-1 protein upstream of Bax/Bak activation. Cells containing a mitochondrial DNA cytochrome b 4-base-pair deletion ([rho−] cells) and cells depleted of their entire mitochondrial DNA ([rho0] cells) are oxidative phosphorylation incompetent and displayed loss of the Mcl-1 protein under anoxia. [rho0] cells, in contrast to [rho−] cells, did not die under anoxia. However, [rho0] cells did undergo cell death in the presence of the Bad BH3 peptide, an inhibitor of Bcl-XL/Bcl-2 proteins. These results indicate that [rho0] cells survive under anoxia despite the loss of Mcl-1 protein due to residual prosurvival activity of the Bcl-XL/Bcl-2 proteins. Collectively, these results demonstrate that anoxia-induced cell death requires the loss of Mcl-1 protein and inhibition of the electron transport chain to negate Bcl-XL/Bcl-2 proteins.


Cell Cycle | 2007

Role of Bcl-2 family members in anoxia induced cell death.

Emelyn H. Shroff; Colleen Snyder; Navdeep S. Chandel

Anoxia, the condition of oxygen deprivation, induces apoptosis via the intrinsic apoptoticpathway. Cells deficient in both Bax and Bak do not undergo cell death during anoxia.However, the underlying mechanism of anoxia induced cell death is not well defined. Herewe report our latest findings of two critical events that are required to induce cell deathduring anoxia. First, a key member of the Bcl-2 family of pro-survival proteins, Mcl-1,undergoes proteasomal-dependent degradation. The loss of Mcl-1 protein is independentof Bax or Bak indicating this is an early event in the apoptotic cascade. Second, cellsinhibit the mitochondrial electron transport chain to negate the pro-survival function of Bcl-2/Bcl-XL. These observations indicate that loss of pro-survival function is necessary foranoxia induced cell death.


PLOS ONE | 2009

BH3 Peptides Induce Mitochondrial Fission and Cell Death Independent of BAX/BAK

Emelyn H. Shroff; Colleen Snyder; G. R. Scott Budinger; Manu Jain; Teng Leong Chew; Satya Khuon; Harris Perlman; Navdeep S. Chandel

BH3 only proteins trigger cell death by interacting with pro- and anti-apoptotic members of the BCL-2 family of proteins. Here we report that BH3 peptides corresponding to the death domain of BH3-only proteins, which bind all the pro-survival BCL-2 family proteins, induce cell death in the absence of BAX and BAK. The BH3 peptides did not cause the release of cytochrome c from isolated mitochondria or from mitochondria in cells. However, the BH3 peptides did cause a decrease in mitochondrial membrane potential but did not induce the opening of the mitochondrial permeability transition pore. Interestingly, the BH3 peptides induced mitochondria to undergo fission in the absence of BAX and BAK. The binding of BCL-XL with dynamin-related protein 1 (DRP1), a GTPase known to regulate mitochondrial fission, increased in the presence of BH3 peptides. These results suggest that pro-survival BCL-2 proteins regulate mitochondrial fission and cell death in the absence of BAX and BAK.


Cancer Research | 2005

Two Functional Epitopes of Pigment Epithelial–Derived Factor Block Angiogenesis and Induce Differentiation in Prostate Cancer

Stephanie Filleur; Karl Volz; Thomas Nelius; Yelena Mirochnik; Hanhua Huang; Tetiana Zaichuk; Maria S. Aymerich; Sofia P. Becerra; Ronald L. Yap; Dorina Veliceasa; Emelyn H. Shroff; Olga V. Volpert


Molecular Cancer Research | 2004

Peroxisome proliferator-activated receptor γ ligands improve the antitumor efficacy of thrombospondin peptide ABT510

Hanhua Huang; Steven C. Campbell; Dhugal F. Bedford; Thomas Nelius; Dorina Veliceasa; Emelyn H. Shroff; Jack Henkin; Andrew J. Schneider; Noel P. Bouck; Olga V. Volpert


Antioxidants & Redox Signaling | 2007

Bcl-2 Family Members Regulate Anoxia-Induced Cell Death

Emelyn H. Shroff; Colleen Snyder; Navdeep S. Chandel

Collaboration


Dive into the Emelyn H. Shroff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arin B. Aurora

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge