Network


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

Hotspot


Dive into the research topics where Himavanth R. Gatla is active.

Publication


Featured researches published by Himavanth R. Gatla.


Journal of Biological Chemistry | 2014

Proteasome inhibition increases recruitment of IκB kinase β (IKKβ), S536P-p65, and transcription factor EGR1 to interleukin-8 (IL-8) promoter, resulting in increased IL-8 production in ovarian cancer cells.

Bipradeb Singha; Himavanth R. Gatla; Subrata Manna; Tzu-Pei Chang; Shannon Sanacora; Vladimir Poltoratsky; Ales Vancura; Ivana Vancurova

Background: IL-8 promotes angiogenesis and metastasis in ovarian cancer. Results: Proteasome inhibition induces specific recruitment of IKKβ, EGR-1, and S536P-p65 to the IL-8 promoter. Conclusion: The increased IKKβ, EGR-1, and S536P-p65 recruitment results in the increased IL-8 expression and release in ovarian cancer cells. Significance: The BZ-increased IL-8 release may be responsible for the BZ-limited effectiveness in ovarian cancer treatment. Proinflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8) contributes to ovarian cancer progression through its induction of tumor cell proliferation, survival, angiogenesis, and metastasis. Proteasome inhibition by bortezomib, which has been used as a frontline therapy in multiple myeloma, has shown only limited effectiveness in ovarian cancer and other solid tumors. However, the responsible mechanisms remain elusive. Here, we show that proteasome inhibition dramatically increases the IL-8 expression and release in ovarian cancer cells. The responsible mechanism involves an increased nuclear accumulation of IκB kinase β (IKKβ) and an increased recruitment of the nuclear IKKβ, p65-phosphorylated at Ser-536, and the transcription factor early growth response-1 (EGR-1) to the endogenous IL-8 promoter. Coimmunoprecipitation studies identified the nuclear EGR-1 associated with IKKβ and with p65, with preferential binding to S536P-p65. Both IKKβ activity and EGR-1 expression are required for the increased IL-8 expression induced by proteasome inhibition in ovarian cancer cells. Interestingly, in multiple myeloma cells the IL-8 release is not increased by bortezomib. Together, these data indicate that the increased IL-8 release may represent one of the underlying mechanisms responsible for the decreased effectiveness of proteasome inhibition in ovarian cancer treatment and identify IKKβ and EGR-1 as potential new targets in ovarian cancer combination therapies.


Journal of Immunology | 2013

Proteasome Inhibition by Bortezomib Increases IL-8 Expression in Androgen-Independent Prostate Cancer Cells: The Role of IKKα

Subrata Manna; Bipradeb Singha; Sai Phyo; Himavanth R. Gatla; Tzu-Pei Chang; Shannon Sanacora; Sitharam Ramaswami; Ivana Vancurova

Expression of the proinflammatory and proangiogenic chemokine IL-8, which is regulated at the transcriptional level by NF-κB, is constitutively increased in androgen-independent metastatic prostate cancer and correlates with poor prognosis. Inhibition of NF-κB–dependent transcription was used as an anticancer strategy for the development of the first clinically approved 26S proteasome inhibitor, bortezomib (BZ). Even though BZ has shown remarkable antitumor activity in hematological malignancies, it has been less effective in prostate cancer and other solid tumors; however, the mechanisms have not been fully understood. In this article, we report that proteasome inhibition by BZ unexpectedly increases IL-8 expression in androgen-independent prostate cancer PC3 and DU145 cells, whereas expression of other NF-κB–regulated genes is inhibited or unchanged. The BZ-increased IL-8 expression is associated with increased in vitro p65 NF-κB DNA binding activity and p65 recruitment to the endogenous IL-8 promoter. In addition, proteasome inhibition induces a nuclear accumulation of IκB kinase (IKK)α, and inhibition of IKKα enzymatic activity significantly attenuates the BZ-induced p65 recruitment to IL-8 promoter and IL-8 expression, demonstrating that the induced IL-8 expression is mediated, at least partly, by IKKα. Together, these data provide the first evidence, to our knowledge, for the gene-specific increase of IL-8 expression by proteasome inhibition in prostate cancer cells and suggest that targeting both IKKα and the proteasome may increase BZ effectiveness in treatment of androgen-independent prostate cancer.


Journal of Biological Chemistry | 2017

Histone Deacetylase (HDAC) Inhibition Induces IκB Kinase (IKK)-dependent Interleukin-8/CXCL8 Expression in Ovarian Cancer Cells

Himavanth R. Gatla; Yue Zou; Mohammad M. Uddin; Bipradeb Singha; Pengli Bu; Ales Vancura; Ivana Vancurova

Overexpression of the pro-angiogenic chemokine IL-8 (CXCL8) is associated with a poor prognosis in several solid tumors, including epithelial ovarian cancer (EOC). Even though histone deacetylase (HDAC) inhibition has shown remarkable antitumor activity in hematological malignancies, it has been less effective in solid tumors, including EOC. Here we report results that may explain the decreased efficiency of HDAC inhibition in EOC, based on our data demonstrating that HDAC inhibition specifically induces expression of IL-8/CXCL8 in SKOV3, CAOV3, and OVCAR3 cells. Suppression or neutralization of vorinostat-induced IL-8/CXCL8 potentiates the vorinostat inhibitory effect on cell viability and proliferation. The IL-8/CXCL8 expression induced by vorinostat in EOC cells is dependent on IκB kinase (IKK) activity and associated with a gene-specific recruitment of IKKβ and IKK-dependent recruitment of p65 NFκB to the IL-8/CXCL8 promoter. In addition, HDAC inhibition induces acetylation of p65 and histone H3 and their IL-8/CXCL8 promoter occupancy. In vivo results demonstrate that combining vorinostat and the IKK inhibitor Bay 117085 significantly reduces tumor growth in nude mice compared with control untreated mice or either drug alone. Mice in the combination group had the lowest IL-8/CXCL8 tumor levels and the lowest tumor expression of the murine neutrophil [7/4] antigen, indicating reduced neutrophil infiltration. Together, our results demonstrate that HDAC inhibition specifically induces IL-8/CXCL8 expression in EOC cells and that the mechanism involves IKK, suggesting that using IKK inhibitors may increase the effectiveness of HDAC inhibitors when treating ovarian cancer and other solid tumors characterized by increased IL-8/CXCL8 expression.


Biomolecules | 2015

Transcriptional regulation of chemokine expression in ovarian cancer.

Bipradeb Singha; Himavanth R. Gatla; Ivana Vancurova

The increased expression of pro-inflammatory and pro-angiogenic chemokines contributes to ovarian cancer progression through the induction of tumor cell proliferation, survival, angiogenesis, and metastasis. The substantial potential of these chemokines to facilitate the progression and metastasis of ovarian cancer underscores the need for their stringent transcriptional regulation. In this Review, we highlight the key mechanisms that regulate the transcription of pro-inflammatory chemokines in ovarian cancer cells, and that have important roles in controlling ovarian cancer progression. We further discuss the potential mechanisms underlying the increased chemokine expression in drug resistance, along with our perspective for future studies.


Oncotarget | 2017

Epigenetic regulation of interleukin-8 expression by class I HDAC and CBP in ovarian cancer cells

Himavanth R. Gatla; Yue Zou; Mohammad M. Uddin; Ivana Vancurova

Although inhibitors of epigenetic regulators have been effective in the treatment of cutaneous T cell lymphoma (CTCL) and other hematopoietic malignancies, they have been less effective in solid tumors, including ovarian cancer (OC). We have previously shown that inhibition of histone deacetylase (HDAC) activity induces expression of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (CXCL8, IL-8) in OC cells, resulting in their increased survival and proliferation. Here, we show that in addition to ovarian cancer SKOV3, OVCAR3, and CAOV3 cells, HDAC inhibition induces the CXCL8 expression in HeLa cells, but not in CTCL Hut-78 cells. In OC cells, the CXCL8 expression is induced by pharmacological inhibition of class I HDACs. Interestingly, while an individual suppression of HDAC1, HDAC2, or HDAC3 by corresponding siRNAs inhibits the CXCL8 expression, their simultaneous suppression induces the CXCL8 expression. The induced CXCL8 expression in OC cells is dependent on histone acetyltransferase (HAT) activity of CREB-binding protein (CBP), but not p300, and is associated with HAT-dependent p65 recruitment to CXCL8 promoter. Together, our results show that the CXCL8 expression in OC cells is induced by combined inhibition of HDAC1, -2, and -3, and silenced by suppression of HAT activity of CBP. In addition, our data indicate that the induced CXCL8 expression may be responsible for the limited effectiveness of HDAC inhibitors in OC and perhaps other solid cancers characterized by CXCL8 overexpression, and suggest that targeting class I HDACs and CBP may provide novel combination strategies by limiting the induced CXCL8 expression.


Methods of Molecular Biology | 2014

Evaluating Cytoplasmic and Nuclear Levels of Inflammatory Cytokines in Cancer Cells by Western Blotting

Himavanth R. Gatla; Bipradeb Singha; Valerie Persaud; Ivana Vancurova

Increased expression and cellular release of inflammatory cytokines, interleukin-8 (IL-8; CXCL8), and high mobility group box-1 (HMGB1) are associated with increased cell proliferation, angiogenesis, and metastasis during cancer progression. In prostate and ovarian cancer cells, increased levels of IL-8 and HMGB1 correlate with poor prognosis. We have recently shown that proteasome inhibition by bortezomib (BZ) specifically increases IL-8 release from metastatic prostate and ovarian cancer cells. In this chapter, we describe a protocol to analyze the cytoplasmic and nuclear levels of IL-8 and HMGB1 in prostate and ovarian cancer cells by western blotting. IL-8 is localized in the cytoplasm in both cell types, and its protein levels are significantly increased by BZ. In contrast, HMGB1 is localized in the nucleus, and BZ increases its nuclear levels only in ovarian cancer cells. The protocol includes isolation of cytoplasmic and nuclear extracts, followed by SDS electrophoresis and western blotting, and can be easily modified to analyze the cytoplasmic and nuclear cytokine levels in other cell types.


Oncotarget | 2017

HDAC/IKK inhibition therapies in solid tumors

Ivana Vancurova; Himavanth R. Gatla; Ales Vancura

Increased expression of the pro-angiogenic chemokine interleukin-8 (CXCL8, IL-8) contributes to the progression of solid cancers through its induction of tumor cell survival, proliferation, and metastasis. While histone deacetylase (HDAC) inhibitors have been remarkably effective in treating hematological malignancies, they have been far less effective as single agents in the treatment of solid tumors. We have recently reported results that may explain the limited efficiency of HDAC inhibitors in epithelial ovarian cancer (EOC), based on our data demonstrating that HDAC inhibition induces expression of IL-8 in EOC cells, resulting in their increased viability and proliferation, and that this is dependent on IκB kinase (IKK) activity [1]. Importantly, our in vivo results have demonstrated that combining HDAC and IKK inhibitors significantly reduces ovarian tumor growth when compared to either drug alone [1]. These data provide the first in vivo evidence demonstrating that IKK inhibition increases effectiveness of HDAC inhibitors in suppressing solid tumor growth, and suggesting that using IKK inhibitors may increase effectiveness of HDAC inhibitors in treating ovarian cancer and other solid tumors characterized by increased IL-8 expression (Figure 1). Even though HDAC expression is increased in many types of solid tumors, including EOC, clinical trials targeting the HDAC activity in solid tumors have been disappointing [2, 3]. The rationale for developing HDAC inhibitors as anti-cancer agents was based on their ability to induce hyperacetylation of histones and nonhistone proteins, resulting in increased differentiation, apoptosis, and cell cycle arrest of cancer cells [3, 4]. Vorinostat (suberoylanilide hydroxamic acid, Zolinza) and romidepsin (depsipeptide, FK228, Istodax) are the first HDACs inhibitors that have been approved by the FDA for the treatment of cutaneous T cell lymphoma. However, accumulating evidence indicates that HDAC inhibitors also activate the NFκB pro-survival pathway, which limits their lethality and contributes to drug resistance [5]. Interestingly, we have found that among the tested NFκB-regulated genes, only IL-8 is significantly induced by HDAC inhibition in EOC cells [1]. The induced IL-8 expression is associated with a gene specific, IKKdependent p65 NFκB recruitment to the IL-8 promoter. However, why HDAC inhibition induces p65 recruitment only to the IL-8 promoter in EOC cells? One possible scenario is that in EOC cells, the IL-8 promoter is Editorial


PLOS ONE | 2018

Proteasome inhibition induces IKK-dependent interleukin-8 expression in triple negative breast cancer cells: Opportunity for combination therapy

Mohammad M. Uddin; Yue Zou; Tamanna Sharma; Himavanth R. Gatla; Ivana Vancurova

Triple negative breast cancer (TNBC) cells express increased levels of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8), which promotes their proliferation and migration. Because TNBC patients are unresponsive to current targeted therapies, new therapeutic strategies are urgently needed. While proteasome inhibition by bortezomib (BZ) or carfilzomib (CZ) has been effective in treating hematological malignancies, it has been less effective in solid tumors, including TNBC, but the mechanisms are incompletely understood. Here we report that proteasome inhibition significantly increases expression of IL-8, and its receptors CXCR1 and CXCR2, in TNBC cells. Suppression or neutralization of the BZ-induced IL-8 potentiates the BZ cytotoxic and anti-proliferative effect in TNBC cells. The IL-8 expression induced by proteasome inhibition in TNBC cells is mediated by IκB kinase (IKK), increased nuclear accumulation of p65 NFκB, and by IKK-dependent p65 recruitment to IL-8 promoter. Importantly, inhibition of IKK activity significantly decreases proliferation, migration, and invasion of BZ-treated TNBC cells. These data provide the first evidence demonstrating that proteasome inhibition increases the IL-8 signaling in TNBC cells, and suggesting that IKK inhibitors may increase effectiveness of proteasome inhibitors in treating TNBC.


Methods of Molecular Biology | 2014

Quantitative analysis of bortezomib-induced IL-8 gene expression in ovarian cancer cells.

Bipradeb Singha; Sai Phyo; Himavanth R. Gatla; Ivana Vancurova

Interleukin-8 (IL-8), originally discovered as the neutrophil chemoattractant and inducer of leukocyte-mediated inflammation, contributes to cancer progression through its induction of tumor cell proliferation, survival, and migration. IL-8 expression is increased in many types of advanced cancers, including ovarian cancer, and correlates with poor prognosis. Bortezomib (BZ) is the first FDA-approved proteasome inhibitor that has shown remarkable antitumor activity in multiple myeloma and other hematological malignancies. In solid tumors, including ovarian carcinoma, BZ has been less effective as a single agent; however, the mechanisms remain unknown. We have recently shown that in ovarian cancer cells, BZ greatly increases IL-8 expression, while expression of other NFκB-regulated cytokines, IL-6 and TNF, is unchanged. In this chapter, we describe a protocol that uses real-time qRT-PCR to quantitatively analyze mRNA levels of IL-8 and IL-6 in BZ-treated ovarian cancer cells. The protocol can be easily modified and used for analysis of other cytokines in different cell types.


Oncotarget | 2015

IKK inhibition increases bortezomib effectiveness in ovarian cancer

Bipradeb Singha; Himavanth R. Gatla; Sai Phyo; Atish Patel; Zhe-Sheng Chen; Ivana Vancurova

Collaboration


Dive into the Himavanth R. Gatla's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sai Phyo

St. John's University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yue Zou

St. John's University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge