Network


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

Hotspot


Dive into the research topics where Suryavathi Viswanadhapalli is active.

Publication


Featured researches published by Suryavathi Viswanadhapalli.


Circulation | 2015

NADPH Oxidase 4 Induces Cardiac Fibrosis and Hypertrophy through Activating Akt/mTOR and NFκB Signaling Pathways

Qingwei David Zhao; Suryavathi Viswanadhapalli; Paul J. Williams; Qian Shi; Chunyan Tan; Xiaolan Yi; Basant Bhandari; Hanna E. Abboud

Background— NADPH oxidase 4 (Nox4) has been implicated in cardiac remodeling, but its precise role in cardiac injury remains controversial. Furthermore, little is known about the downstream effector signaling pathways activated by Nox4-derived reactive oxygen species in the myocardium. We investigated the role of Nox4 and Nox4-associated signaling pathways in the development of cardiac remodeling. Methods and Results— Cardiac-specific human Nox4 transgenic mice (c-hNox4Tg) were generated. Four groups of mice were studied: (1) control mice, littermates that are negative for hNox4 transgene but Cre positive; (2) c-hNox4 Tg mice; (3) angiotensin II (AngII)–infused control mice; and (4) c-hNox4Tg mice infused with AngII. The c-hNox4Tg mice exhibited an ≈10-fold increase in Nox4 protein expression and an 8-fold increase in the production of reactive oxygen species, and manifested cardiac interstitial fibrosis. AngII infusion to control mice increased cardiac Nox4 expression and induced fibrosis and hypertrophy. The Tg mice receiving AngII exhibited more advanced cardiac remodeling and robust elevation in Nox4 expression, indicating that AngII worsens cardiac injury, at least in part by enhancing Nox4 expression. Moreover, hNox4 transgene and AngII infusion induced the expression of cardiac fetal genes and activated the Akt-mTOR and NF&kgr;B signaling pathways. Treatment of AngII-infused c-hNox4Tg mice with GKT137831, a Nox4/Nox1 inhibitor, abolished the increase in oxidative stress, suppressed the Akt-mTOR and NF&kgr;B signaling pathways, and attenuated cardiac remodeling. Conclusions— Upregulation of Nox4 in the myocardium causes cardiac remodeling through activating Akt-mTOR and NF&kgr;B signaling pathways. Inhibition of Nox4 has therapeutic potential to treat cardiac remodeling.


Oncogene | 2017

Novel KDM1A inhibitors induce differentiation and apoptosis of glioma stem cells via unfolded protein response pathway

Gangadhara Reddy Sareddy; Suryavathi Viswanadhapalli; P Surapaneni; Takayoshi Suzuki; Andrew Brenner; Ratna K. Vadlamudi

Glioma stem cells (GSCs) have a central role in glioblastoma (GBM) development and chemo/radiation resistance, and their elimination is critical for the development of efficient therapeutic strategies. Recently, we showed that lysine demethylase KDM1A is overexpressed in GBM. In the present study, we determined whether KDM1A modulates GSCs stemness and differentiation and tested the utility of two novel KDM1A-specific inhibitors (NCL-1 and NCD-38) to promote differentiation and apoptosis of GSCs. The efficacy of KDM1A targeting drugs was tested on purified GSCs isolated from established and patient-derived GBMs using both in vitro assays and in vivo orthotopic preclinical models. Our results suggested that KDM1A is highly expressed in GSCs and knockdown of KDM1A using shRNA-reduced GSCs stemness and induced the differentiation. Pharmacological inhibition of KDM1A using NCL-1 and NCD-38 significantly reduced the cell viability, neurosphere formation and induced apoptosis of GSCs with little effect on differentiated cells. In preclinical studies using orthotopic models, NCL-1 and NCD-38 significantly reduced GSCs-driven tumor progression and improved mice survival. RNA-sequencing analysis showed that KDM1A inhibitors modulate several pathways related to stemness, differentiation and apoptosis. Mechanistic studies showed that KDM1A inhibitors induce activation of the unfolded protein response (UPR) pathway. These results strongly suggest that selective targeting of KDM1A using NCL-1 and NCD-38 is a promising therapeutic strategy for elimination of GSCs.


Scientific Reports | 2016

Selective Estrogen Receptor β Agonist LY500307 as a Novel Therapeutic Agent for Glioblastoma

Gangadhara Reddy Sareddy; Xiaonan Li; Jinyou Liu; Suryavathi Viswanadhapalli; Lauren Garcia; Aleksandra Gruslova; David Cavazos; Mike Garcia; Anders Ström; Jan Åke Gustafsson; Rajeshwar Rao Tekmal; Andrew Brenner; Ratna K. Vadlamudi

Glioblastomas (GBM), deadly brain tumors, have greater incidence in males than females. Epidemiological evidence supports a tumor suppressive role of estrogen; however, estrogen as a potential therapy for GBM is limited due to safety concerns. Since GBM express ERβ, a second receptor for estrogen, targeting ERβ with a selective agonist may be a potential novel GBM therapy. In the present study, we examined the therapeutic effect of the selective synthetic ERβ agonist LY500307 using in vitro and in vivo GBM models. Treatment with LY500307 significantly reduced the proliferation of GBM cells with no activity on normal astrocytes in vitro. ERβ agonists promoted apoptosis of GBM cells, and mechanistic studies using RNA sequencing revealed that LY500307 modulated several pathways related to apoptosis, cell cycle, and DNA damage response. Further, LY500307 sensitized GBM cells to several FDA-approved chemotherapeutic drugs including cisplatin, lomustine and temozolomide. LY500307 treatment significantly reduced the in vivo tumor growth and promoted apoptosis of GBM tumors in an orthotopic model and improved the overall survival of tumor-bearing mice in the GL26 syngeneic glioma model. Our results demonstrate that LY500307 has potential as a therapeutic agent for GBM.


eLife | 2017

Estrogen receptor coregulator binding modulators (ERXs) effectively target estrogen receptor positive human breast cancers

Ganesh V. Raj; Gangadhara Reddy Sareddy; Shihong Ma; Tae-Kyung Lee; Suryavathi Viswanadhapalli; Rui Li; Xihui Liu; Shino Murakami; Chien Cheng Chen; Wan Ru Lee; Monica Mann; Samaya Rajeshwari Krishnan; Bikash Manandhar; Vijay K. Gonugunta; Douglas W. Strand; Rajeshwar Rao Tekmal; Jung Mo Ahn; Ratna K. Vadlamudi

The majority of human breast cancer is estrogen receptor alpha (ER) positive. While anti-estrogens/aromatase inhibitors are initially effective, resistance to these drugs commonly develops. Therapy-resistant tumors often retain ER signaling, via interaction with critical oncogenic coregulator proteins. To address these mechanisms of resistance, we have developed a novel ER coregulator binding modulator, ERX-11. ERX-11 interacts directly with ER and blocks the interaction between a subset of coregulators with both native and mutant forms of ER. ERX-11 effectively blocks ER-mediated oncogenic signaling and has potent anti-proliferative activity against therapy-sensitive and therapy-resistant human breast cancer cells. ERX-11 is orally bioavailable, with no overt signs of toxicity and potent activity in both murine xenograft and patient-derived breast tumor explant models. This first-in-class agent, with its novel mechanism of action of disrupting critical protein-protein interactions, overcomes the limitations of current therapies and may be clinically translatable for patients with therapy-sensitive and therapy-resistant breast cancers. DOI: http://dx.doi.org/10.7554/eLife.26857.001


Nature Communications | 2017

Cross-talk between miR-471-5p and autophagy component proteins regulates LC3-associated phagocytosis (LAP) of apoptotic germ cells

Subbarayalu Panneerdoss; Suryavathi Viswanadhapalli; Nourhan Abdelfattah; Benjamin Onyeagucha; Santosh Timilsina; Tabrez A. Mohammad; Yidong Chen; Michael T. Drake; Kristiina Vuori; T. Rajendra Kumar; Manjeet K. Rao

Phagocytic clearance of apoptotic germ cells by Sertoli cells is vital for germ cell development and differentiation. Here, using a tissue-specific miRNA transgenic mouse model, we show that interaction between miR-471-5p and autophagy member proteins regulates clearance of apoptotic germ cells via LC3-associated phagocytosis (LAP). Transgenic mice expressing miR-471-5p in Sertoli cells show increased germ cell apoptosis and compromised male fertility. Those effects are due to defective engulfment and impaired LAP-mediated clearance of apoptotic germ cells as miR-471-5p transgenic mice show lower levels of Dock180, LC3, Atg12, Becn1, Rab5 and Rubicon in Sertoli cells. Our results reveal that Dock180 interacts with autophagy member proteins to constitute a functional LC3-dependent phagocytic complex. We find that androgen regulates Sertoli cell phagocytosis by controlling expression of miR-471-5p and its target proteins. These findings suggest that recruitment of autophagy machinery is essential for efficient clearance of apoptotic germ cells by Sertoli cells using LAP.Although phagocytic clearance of apoptotic germ cells by Sertoli cells is essential for spermatogenesis, little of the mechanism is known. Here the authors show that Sertoli cells employ LC3-associated phagocytosis (LAP) by recruiting autophagy member proteins to clear apoptotic germ cells.


Oncotarget | 2017

Therapeutic utility of natural estrogen receptor beta agonists on ovarian cancer

Jinyou Liu; Suryavathi Viswanadhapalli; Lauren Garcia; Mei Zhou; Binoj C. Nair; Edward R. Kost; Rajeshwar Rao Tekmal; Rong Li; Manjeet K. Rao; Tyler J. Curiel; Ratna K. Vadlamudi; Gangadhara Reddy Sareddy

Ovarian cancer is the deadliest of all gynecologic cancers. Despite success with initial chemotherapy, the majority of patients relapse with an incurable disease. Development of chemotherapy resistance is a major factor for poor long-term survival in ovarian cancer. The biological effects of estrogens are mediated by estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). Emerging evidence suggests that ovarian cancer cells express ERβ that functions as a tumor suppressor; however, the clinical utility of ERβ agonists in ovarian cancer remains elusive. We tested the utility of two natural ERβ agonists liquiritigenin (Liq), which is isolated from Glycyrrhiza uralensis and S-equol, which is isolated from soy isoflavone daidzein, for treating ovarian cancer. Both natural ERβ ligands had significant growth inhibition in cell viability and survival assays, reduced migration and invasion, and promoted apoptosis. Further, ERβ agonists showed tumor suppressive functions in therapy-resistant ovarian cancer model cells and sensitized ovarian cancer cells to cisplatin and paclitaxel treatment. Global RNA-Seq analysis revealed that ERβ agonists modulate several tumor suppressive pathways, including downregulation of the NF-κB pathway. Immunoprecipitation assays revealed that ERβ interacts with p65 subunit of NF-κB and ERβ overexpression reduced the expression of NF-κB target genes. In xenograft assays, ERβ agonists reduced tumor growth and promoted apoptosis. Collectively, our findings demonstrated that natural ERβ agonists have the potential to significantly inhibit ovarian cancer cell growth by anti-inflammatory and pro-apoptotic actions, and natural ERβ agonists represent novel therapeutic agents for the management of ovarian cancer.


Cancer Research | 2017

Abstract 4148: Novel ERX-11 and CDK4/6 inhibitor combination therapy for treating therapy resistant breast cancer

Suryavathi Viswanadhapalli; Gangadhara Reddy Sareddy; Shihong Ma; Tae-Kyung Lee; Rajeshwar Rao Tekmal; Jung Mo Ahn; Ganesh V. Raj; Ratna K. Vadlamudi

BACKGROUND: The majority of the breast cancer is estrogen receptor alpha (ESR1) positive. While tamoxifen and letrozole therapies are effective, therapy resistance is common. Importantly, both therapy-sensitive and therapy-resistant tumors retain ESR1 signaling, via interaction with critical oncogenic coregulator proteins. Further, resistant tumors commonly acquire cyclin D1:CDK4/6 signaling via multiple mechanisms, cyclin D1 can independently activate ESR1 and thus contribute to estrogen independence of ESR+ tumor. Currently, CDK4/6 inhibitors in clinical trials for treating breast cancer, however, considering complex signaling interplay of estrogen and CDK axis, combination therapy of CDK inhibitor with other potent ESR1 targeted agents may have better utility and may prevent development of resistance to the CDK4/6 inhibitors. We recently developed a small organic molecule, ESR1 coregulator binding inhibitor ERX-11 (EtiraRx-11). The objective of this study is to test the utility of novel combination therapy of ERX-11 with CDK4/6 inhibitor palbociclib in treating therapy resistant cancer. METHODS: We have utilized multiple therapy sensitive and therapy-resistant models with various genetic back grounds. We tested efficacy using both acquired resistance and engineered models that express ESR1 mutations or oncogenes. Efficacy of combination therapy was tested using established in vitro assays including, MTT, colony formation, apoptosis, and cell cycle progression. Mechanistic studies were conducted using reporter gene assays, gene expression and signaling alterations. Xenograft studies were used to determine the in vivo efficacy of the combination therapy. RESULTS: ERX-11 effectively blocked ESR1-mediated oncogenic signaling and has potent anti-proliferative activity against therapy-sensitive and therapy-resistant breast cancer cells. Mechansistic studies showed that ERX-11 blocks the interaction between a subset of coregulators with both native and mutant forms of ESR1. ERX-11 showed potent activity in both preclinical xenograft models and patient-derived breast tumor explant models. Co-treatment of ERX-11 with palbociclib synergistically reduced cell viability and induced apoptosis of therapy sensitive and resistant breast cancer model cells. Importantly, combination therapy of ERX-11 and the palbociclib synergistically reduced the growth and induced apoptosis of tamoxifen and letrozole resistant xenograft tumors compared to either drug alone. Mechanistic studies showed combination therapy significantly altered E2F1 and ESR1 signaling pathways and promoted apoptosis. CONCLUSIONS: Collectively our studies have discovered a novel combinational treatment with ERX-11 and palbociclib for patients with therapy-sensitive and therapy-resistant breast cancers. Citation Format: Suryavathi Viswanadhapalli, Gangadhara Reddy Sareddy, Shi-Hong Ma, Tae-Kyung Lee, Rajeshwar Rao Tekmal, Jung-Mo Ahn, Ganesh Raj, Ratna K. Vadlamudi. Novel ERX-11 and CDK4/6 inhibitor combination therapy for treating therapy resistant breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4148. doi:10.1158/1538-7445.AM2017-4148


Scientific Reports | 2018

Nox4 is a Target for Tuberin Deficiency Syndrome

Qian Shi; Suryavathi Viswanadhapalli; William E. Friedrichs; Chakradhar Velagapudi; Cedric Szyndralewiez; Shweta Bansal; Manzoor A. Bhat; Goutam Ghosh Choudhury; Hanna E. Abboud

The mechanism by which TSC2 inactivation or deficiency contributes to the pathology of tuberous sclerosis complex (TSC) is not fully clear. We show that renal angiomyolipomas from TSC patients and kidney cortex from Tsc2+/− mice exhibit elevated levels of reactive oxygen species (ROS). Downregulation of tuberin (protein encoded by TSC2 gene) in renal proximal tubular epithelial cells significantly increased ROS concomitant with enhanced Nox4. Similarly, we found elevated levels of Nox4 in the renal cortex of Tsc2+/− mice and in the renal angiomyolipomas from TSC patients. Tuberin deficiency is associated with activation of mTORC1. Rapamycin, shRNAs targeting raptor, or inhibition of S6 kinase significantly inhibited the expression of Nox4, resulting in attenuation of production of ROS in tuberin-downregulated proximal tubular epithelial cells. In contrast, activation of mTORC1 increased Nox4 and ROS. These results indicate that Nox4 may be a potential target for tuberin-deficiency-derived diseases. Using a xenograft model from tuberin-null tubular cells in nude mice, both anti-sense Nox4 and GKT137831, a specific inhibitor of Nox1/4, significantly inhibited the tumor growth. Thus, our results demonstrate the presence of an antagonistic relationship between tuberin and Nox4 to drive oncogenesis in the tuberin deficiency syndrome and identify Nox4 as a target to develop a therapy for TSC.


Science Advances | 2018

Cross-talk among writers, readers, and erasers of m6A regulates cancer growth and progression

Subbarayalu Panneerdoss; Vijay K. Eedunuri; Pooja Yadav; Santosh Timilsina; Subapriya Rajamanickam; Suryavathi Viswanadhapalli; Nourhan Abdelfattah; Benjamin Onyeagucha; Xiadong Cui; Zhao Lai; Tabrez A. Mohammad; Yogesh K. Gupta; Tim H M Huang; Yufei Huang; Yidong Chen; Manjeet K. Rao

Collaboration among writers-readers-erasers of m6A regulates the stability of tumor-specific genes. The importance of RNA methylation in biological processes is an emerging focus of investigation. We report that altering m6A levels by silencing either N6-adenosine methyltransferase METTL14 (methyltransferase-like 14) or demethylase ALKBH5 (ALKB homolog 5) inhibits cancer growth and invasion. METTL14/ALKBH5 mediate their protumorigenic function by regulating m6A levels of key epithelial-mesenchymal transition and angiogenesis-associated transcripts, including transforming growth factor–β signaling pathway genes. Using MeRIP-seq (methylated RNA immunoprecipitation sequencing) analysis and functional studies, we find that these target genes are particularly sensitive to changes in m6A modifications, as altered m6A status leads to aberrant expression of these genes, resulting in inappropriate cell cycle progression and evasion of apoptosis. Our results reveal that METTL14 and ALKBH5 determine the m6A status of target genes by controlling each other’s expression and by inhibiting m6A reader YTHDF3 (YTH N6-methyladenosine RNA binding protein 3), which blocks RNA demethylase activity. Furthermore, we show that ALKBH5/METTL14 constitute a positive feedback loop with RNA stability factor HuR to regulate the stability of target transcripts. We discover that hypoxia alters the level/activity of writers, erasers, and readers, leading to decreased m6A and consequently increased expression of target transcripts in cancer cells. This study unveils a previously undefined role for m6A in cancer and shows that the collaboration among writers-erasers-readers sets up the m6A threshold to ensure the stability of progrowth/proliferation-specific genes, and protumorigenic stimulus, such as hypoxia, perturbs that m6A threshold, leading to uncontrolled expression/activity of those genes, resulting in tumor growth, angiogenesis, and progression.


Molecular Cancer Research | 2016

Abstract B08: ESR1 coregulator binding site inhibitors (ECBIs) as novel therapeutics to target hormone therapy-resistant breast cancer

Ratna K. Vadlamudi; Gangadhara Reddy Sareddy; Suryavathi Viswanadhapalli; Tae-Kyung Lee; Shihong Ma; Wan Ru Lee; Monica Mann; Samaya Rajeshwari Krishnan; Vijay K. Gonugunta; Douglas W. Strand; Rajeshwar Rao Tekmal; Jung Mo Ahn; Ganesh V. Raj

Estrogens contribute to the progression of breast cancer via estrogen receptor 1 (ESR1) and current therapies involve either antiestrogens (AE) or aromatase inhibitors (AI). However, most patients develop resistance to these drugs. Critically, therapy-resistant tumors retain ESR1-signaling. Mechanisms of therapy resistance involve the activation of ESR1 in the absence of ligand or mutations in ESR1 that allow interaction between the ESR1 and coregulators leading to sustained ESR1 signaling and proliferation. For patients with therapy-resistant breast cancers, there is a critical unmet need for novel agents to disrupt ESR1 signaling by blocking ESR1 interactions with its coregulators. Methods: Using rational design, we synthesized and evaluated a small organic molecule (ESR1 coregulator binding inhibitor, ECBI) that mimics the ESR1 coregulator nuclear receptor box motif. Using in vitro cell proliferation and apoptosis assays, we tested the effect of ECBI on several breast cancer cells and therapy-resistant model cells. Mechanistic studies were conducted using established biochemical assays, reporter gene assays, RTqPCR and RNASeq analysis. Gene differential expression lists were analyzed using Ingenuity Pathway Analysis (IPA). ESR1+ve (MCF7 and ZR75) xenografts were used for preclinical evaluation and toxicity. The efficacy of ECBI was tested using an ex vivo cultures of freshly extirpated prrimary human breast tissues. Results: In estrogen induced proliferation assays using several ESR1+ve model cells, we found that ECBI inhibit growth (IC50=300-500 nM). Importantly, ECBI showed little or no activity on ESR1 negative cells. Further, ECBI also reduced the proliferation of several ESR1 positive hormonal therapy resistant cells, directly interacted with MT-ESR1 with high affinity and significantly inhibited MT-ESR1 driven oncogenic activity. Mechanistic studies showed that ECBI interacts with ESR1, efficiently blocks ESR1 interactions with coregulators and reduces the ESR1 reporter gene activity. RNA sequencing analysis revealed that ECBI blocks multiple ESR1 driven pathways, likely representing the ability of a single ECBI compound to block multiple ESR1-coregulator interactions. Treatment of ESR1-positive xenograft tumors with ECBI (10 mg/Kg/oral) reduced tumor volume by 67% compared to control. Further, ECBI also significantly reduced the proliferation of coregulator-overexpressed breast cancer cells in xenograft model. Using human primary breast tissue ex vivo cultures, we have provided evidence that ECBI has potential to dramatically reduce proliferation of human breast tumor cells. Conclusions: The ECBI is a novel agent that targets ESR1 with a unique mechanism of action. ECBI has distinct pharmacologic advantages of oral bioavailability, in vivo stability, and is associated with minimal systemic side effects. Remarkably, ECBIs block both native and mutant forms of ESR1 and have activity against therapy resistant breast cancer cell proliferation both in vitro and in vivo and against primary human tissues ex vivo. Thus development of ECBI represents a quantum leap in therapies to target ESR1 Citation Format: Ratna K. Vadlamudi, Gangadhara Reddy Sareddy, Suryavathi Viswanadhapalli, Tae-Kyung Lee, Shi-Hong Ma, Wan Ru Lee, Monica Mann, Samaya Rajeshwari Krishnan, Vijay Gonugunta, Douglas W. Strand, Rajeshwar Rao Tekmal, JungMo Ahn, Ganesh V. Raj. ESR1 coregulator binding site inhibitors (ECBIs) as novel therapeutics to target hormone therapy-resistant breast cancer. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research; Oct 17-20, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(2_Suppl):Abstract nr B08.

Collaboration


Dive into the Suryavathi Viswanadhapalli's collaboration.

Top Co-Authors

Avatar

Gangadhara Reddy Sareddy

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Ratna K. Vadlamudi

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Rajeshwar Rao Tekmal

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Andrew Brenner

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Jinyou Liu

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Mei Zhou

Central South University

View shared research outputs
Top Co-Authors

Avatar

Manjeet K. Rao

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Monica Mann

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Ganesh V. Raj

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hanna E. Abboud

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge