Network


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

Hotspot


Dive into the research topics where Indira V. Subramanian is active.

Publication


Featured researches published by Indira V. Subramanian.


Journal of Clinical Investigation | 2010

Hypoxia-induced microRNA-424 expression in human endothelial cells regulates HIF-α isoforms and promotes angiogenesis

Goutam Ghosh; Indira V. Subramanian; Neeta Adhikari; Xiaoxiao Zhang; Hemant Joshi; David L. Basi; Y. Chandrashekhar; Jennifer L. Hall; Sabita Roy; Yan Zeng; Sundaram Ramakrishnan

Adaptive changes to oxygen availability are critical for cell survival and tissue homeostasis. Prolonged oxygen deprivation due to reduced blood flow to cardiac or peripheral tissues can lead to myocardial infarction and peripheral vascular disease, respectively. Mammalian cells respond to hypoxia by modulating oxygen-sensing transducers that stabilize the transcription factor hypoxia-inducible factor 1α (HIF-1α), which transactivates genes governing angiogenesis and metabolic pathways. Oxygen-dependent changes in HIF-1α levels are regulated by proline hydroxylation and proteasomal degradation. Here we provide evidence for what we believe is a novel mechanism regulating HIF-1α levels in isolated human ECs during hypoxia. Hypoxia differentially increased microRNA-424 (miR-424) levels in ECs. miR-424 targeted cullin 2 (CUL2), a scaffolding protein critical to the assembly of the ubiquitin ligase system, thereby stabilizing HIF-α isoforms. Hypoxia-induced miR-424 was regulated by PU.1-dependent transactivation. PU.1 levels were increased in hypoxic endothelium by RUNX-1 and C/EBPα. Furthermore, miR-424 promoted angiogenesis in vitro and in mice, which was blocked by a specific morpholino. The rodent homolog of human miR-424, mu-miR-322, was significantly upregulated in parallel with HIF-1α in experimental models of ischemia. These results suggest that miR-322/424 plays an important physiological role in post-ischemic vascular remodeling and angiogenesis.


Autophagy | 2007

Autophagy and angiogenesis inhibition

Sundaram Ramakrishnan; Tri Minh Bui Nguyen; Indira V. Subramanian; Ameeta Kelekar

Angiogenesis, the process by which new blood vessels are formed is critical for embryonic development and physiological functioning of normal tissues. Angiogenesis also plays a critical role in the pathology of many diseases including cancer, wherein the supply and demand for blood vessels determines the rate of cancer growth. A number of therapeutic strategies are being developed to inhibit pathological angiogenesis. Kringle domains of plasminogen such as kringle 5 (K5) and a proteolytic fragment of collagen type XVIII (endostatin) are well-characterized, potent angiogenesis inhibitors. These inhibitors activate different intracellular signaling pathways to induce apoptosis and inhibit cell proliferation. Recent studies from our group have shown that K5 and endostatin can also induce autophagy in addition to apoptosis in endothelial cells. A common feature of the two treatments was the upregulation of Beclin 1 levels leading to alterations in the Beclin 1-Bcl-2 complex. Angiogenesis inhibitor-induced autophagy in endothelial cells was independent of nutritional or hypoxic stress and initiated even in the presence of endothelial-specific survival factors such as vascular endothelial growth factor (VEGF). Interfering with the autophagic response by knocking down Beclin 1 levels dramatically increased apoptosis of endothelial cells. These findings identify the autophagic response as a novel target for enhancing the therapeutic efficacy of angiogenesis inhibitors. Addendum to: Kringle 5 of Human Plasminogen, an Angiogenesis Inhibitor, Induces Both Autophagy and Apoptotic Death in Endothelial Cells T.M.B. Nguyen, I.V. Subramanian, A. Kelekar and S. Ramakrishnan Blood 2007; 109:4793-802


Journal of Cellular and Molecular Medicine | 2009

Endostatin induces autophagy in endothelial cells by modulating Beclin 1 and β‐catenin levels

Tri Minh Bui Nguyen; Indira V. Subramanian; Xue Xiao; Goutam Ghosh; Phan Nguyen; Ameeta Kelekar; Sundaram Ramakrishnan

Endostatin is a well‐characterized endogenous inhibitor of angiogenesis that affects cell proliferation and migration by inhibiting integrin and Wnt‐mediated signalling pathways. Here, we show that endothelial cells treated with native and P125A‐endostatin activate autophagy. Because autophagy can either be protective or induce programmed cell death, experiments were carried out to understand the signalling pathways leading to autophagy in endothelial cells. P125A‐endostatin treatment increased the levels of Beclin 1, a crucial molecule in vesicle nucleation and autophagy. The treatment also reduced the levels of Bcl‐2, Bcl‐xL and β‐catenin; however, progressively increasing amounts of Bcl‐2 and Bcl‐xL were found to be complexed with Beclin 1. Increased β‐catenin and Wnt‐mediated signalling reduced Beclin 1 levels and rescued endothelial cells from endostatin‐induced autophagy. Finally, knocking down Beclin 1 levels by RNA interference decreased autophagy and accelerated caspase activation in endostatin‐treated cells. These studies suggest that endothelial cells may initiate autophagy as a survival response to limit the effects of angiogenesis inhibitors. Thus, interfering with autophagy can potentiate the effects of endostatin by promoting a switch to apoptosis.


International Journal of Cancer | 2004

Carboplatin selectively induces the VEGF stress response in endothelial cells: Potentiation of antitumor activity by combination treatment with antibody to VEGF.

Robert Wild; Ruud P.M. Dings; Indira V. Subramanian; Sundaram Ramakrishnan

Vascular Endothelial Growth Factor (VEGF) functions as a key regulator in tumor angiogenesis. In addition, VEGF is an important survival factor for endothelial cells under chemical or physical stress. In our report, we show that treatment of endothelial cells with the chemotherapeutic agent carboplatin significantly increased the expression of VEGF. Furthermore, neutralization of secreted VEGF with specific polyclonal anti‐VEGF antibodies or monoclonal antibody sensitized endothelial cells to carboplatin treatment and increased apoptosis several‐fold. Interestingly, carboplatin treatment did not alter VEGF expression in tumor cells. Similarly, antibody to VEGF did not change the chemosensitivity of tumor cells to this drug. Most importantly, tumor‐bearing animals treated with carboplatin showed an increase in VEGF immunoreactivity in the tumor vasculature, confirming the in vitro studies. Based on these observations, we determined whether neutralization of VEGF could enhance the anti‐tumor activity of carboplatin in an in vivo ovarian cancer model system. A combination therapy consisting of a suboptimal dose of carboplatin (32.5 mg/kg/inj., q3d×5; i.p.) and polyclonal anti‐VEGF antibody (2 mg/inj., q3d×10; i.p.) significantly enhanced solid tumor growth inhibition over individual monotherapies and included multiple complete responses. These findings suggest that VEGF is a critical endothelial cell specific survival factor that is induced by carboplatin and contributes to the protection of tumor vasculature during chemotherapy treatment. In addition, these results provide evidence for a potential mechanism that underlies enhanced anti‐tumor activity achieved with chemotherapy and anti‐VEGF antibody combination treatment regimens as recently reported in a number of clinical trials. We conclude that a similar type of combination therapy may be applicable to many types of malignancies since VEGF expression was differentially induced in the tumor host environment (i.e., tumor vasculature) and not in the tumor cells themselves; hence, this phenomenon may be independent of the type and origin of the primary cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Dynamin 2 along with microRNA-199a reciprocally regulate hypoxia-inducible factors and ovarian cancer metastasis

Hemant Joshi; Indira V. Subramanian; Erica Schnettler; Goutam Ghosh; Rajesha Rupaimoole; C. Evans; Manju Saluja; Yawu Jing; Ivan Cristina; Sabita Roy; Yan Zeng; Vijay H. Shah; Anil K. Sood; Sundaram Ramakrishnan

Significance Tumor cells adapt to hypoxia by modulating energy production and utilization. Endocytosis is an energy-consuming process that is suppressed during hypoxia. Our studies show that Dynamin 2 (DNM2), a key component of endocytic machinery, is transcriptionally suppressed by HIF-1. Genetic and pharmacological inactivation of DNM2 stabilized HIF-1α and HIF-2α. Furthermore, miR-199a, which is encoded from the opposite strand of DNM2, exerts reciprocal negative regulation upon HIF-1α and HIF-2α. Overexpression of miR-199a decreased HIF-1α and HIF-2α, cell migration, and metastasis. These findings establish a regulatory loop between endocytic pathway and hypoxic response in tumor cells. Hypoxia-driven changes in the tumor microenvironment facilitate cancer metastasis. In the present study, we investigated the regulatory cross talk between endocytic pathway, hypoxia, and tumor metastasis. Dynamin 2 (DNM2), a GTPase, is a critical mediator of endocytosis. Hypoxia decreased the levels of DNM2. DNM2 promoter has multiple hypoxia-inducible factor (HIF)-binding sites and genetic deletion of them relieved hypoxia-induced transcriptional suppression. Interestingly, DNM2 reciprocally regulated HIF. Inhibition of DNM2 GTPase activity and dominant-negative mutant of DNM2 showed a functional role for DNM2 in regulating HIF. Furthermore, the opposite strand of DNM2 gene encodes miR-199a, which is similarly reduced in cancer cells under hypoxia. miR-199a targets the 3′-UTR of HIF-1α and HIF-2α. Decreased miR-199a expression in hypoxia increased HIF levels. Exogenous expression of miR-199a decreased HIF, cell migration, and metastasis of ovarian cancer cells. miR-199a–mediated changes in HIF levels affected expression of the matrix-remodeling enzyme, lysyloxidase (LOX). LOX levels negatively correlated with progression-free survival in ovarian cancer patients. These results demonstrate a regulatory relationship between DNM2, miR-199a, and HIF, with implications in cancer metastasis.


Cancer Research | 2007

Modulation of Angiogenic Phenotype Alters Tumorigenicity in Rat Ovarian Epithelial Cells

Jennifer J. Schumacher; Ruud P.M. Dings; Jonathan A. Cosin; Indira V. Subramanian; Nelly Auersperg; Sundaram Ramakrishnan

Vascular endothelial growth factor (VEGF) expression correlates with microvessel density, stage, malignant ascites, metastasis, and survival in ovarian cancer. By transducing VEGF165 into a nontumorigenic rat ovarian surface epithelial cell line (ROSE199), we investigated the direct effect of an angiogenic phenotype on tumor development. The neu oncogene, which is overexpressed in >30% of ovarian cancers, was used in comparison. Neu-transfected ROSE199 cells showed phenotypic characteristics of transformation in vitro with an abundance of focus-forming units in monolayer cultures and anchorage-independent growth in soft agar. In contrast, VEGF-secreting ROSE199 cells (VR) retained normal morphology and in vitro growth characteristics (e.g., proliferation rate) compared with parental ROSE199 cells. Interestingly, injection of VR cells into athymic mice formed malignant ascites in 100% of the animals when injected into the peritoneum and developed vascularized tumors in 85% of the mice when injected s.c. Furthermore, blocking VEGF-mediated signaling by the Flk-1/KDR receptor kinase inhibitor SU5416 significantly inhibited the growth of VR tumors. To validate that the proangiogenic switch is responsible for tumor development, the angiogenic phenotype was balanced by the inducible coexpression of endostatin under the control of Tet-activated promoter. Coexpression of endostatin along with VEGF reversed the tumorigenic phenotype of VR cells. These studies show that alterations in the angiogenic characteristics of ovarian surface epithelium may play an important role in the etiology of ovarian cancer, and that inhibition of angiogenesis can be effective in the treatment of epithelial ovarian cancer.


International Journal of Cancer | 2011

Inhibition of ovarian cancer by RGD-P125A-endostatin-Fc fusion proteins

Yawu Jing; Huarui Lu; Kailang Wu; Indira V. Subramanian; Sundaram Ramakrishnan

Previous studies have shown that a single point mutation in endostatin at position 125 (P125A) can improve the biological activity of endostatin. Addition of an integrin‐targeting moiety, R‐G‐D, resulted in better localization to tumor vasculature and improved the antiangiogenic activity of endostatin. Because endostatin has relatively shorter serum half‐life, frequent dosing was required for inhibiting tumor growth. In our study, we have genetically fused RGD‐P125A‐endostatin to Fc of IgG4 isotype and evaluated its antiangiogenic and antitumor effects in athymic mice. Two genetic constructs were made, RGD‐P125A‐endostatin‐Fc (RE‐Fc) and P125A‐endostatin‐RGD‐Fc (ER‐Fc). Both constructs were cloned and expressed in mammalian cells. Purified fusion proteins inhibited endothelial cell migration and proliferation better than yeast‐derived P125A‐endostatin. Both RE‐Fc and ER‐Fc inhibited ovarian cancer growth and were found to be as effective as Bevacizumab treatment. Fusion protein showed marked increased half‐life. Combination treatment with Bevacizumab and ER‐Fc showed additive inhibition of ovarian cancer growth. These studies demonstrate that genetic fusion with human IgG4‐Fc increases the half‐life of P125A‐endostatin and can be used along with Bevacizumab to improve antiangiogenic and antitumor activities.


Gene Therapy | 2010

Adeno-associated virus-mediated delivery of kringle 5 of human plasminogen inhibits orthotopic growth of ovarian cancer

T. M B Nguyen; Indira V. Subramanian; X. Xiao; P. Nguyen; Sundaram Ramakrishnan

Kringle 5 (K5) of human plasminogen is a potent angiogenesis inhibitor. In this study, we investigated the effects of recombinant adeno-associated virus (AAV)-mediated delivery of K5 in mouse models of human ovarian cancer. A single intramuscular injection of AAV-K5 resulted in sustained expression of K5 reaching a maximum serum level of 800 ng ml−1. Gene therapy inhibited both vascular endothelial growth factor (VEGF)-induced and tumor cell-induced angiogenesis in matrigel plug assays. Furthermore, a single injection of AAV-K5 significantly inhibited both subcutaneous and intraperitoneal growth of human ovarian cancer cells. Immunofluorescence studies of residual tumors surgically resected from the treated animals showed reduced tumor burden, which correlated with the inhibition of tumor neovascularization. In addition, AAV-K5 gene therapy differentially affected the nascent vessels more than mature vasculature and induced apoptotic death of tumor cells. These data show that AAV-K5 can be effectively used to inhibit ovarian cancer.


Gene Therapy | 2011

AAV-P125A-endostatin and paclitaxel treatment increases endoreduplication in endothelial cells and inhibits metastasis of breast cancer

Indira V. Subramanian; S. Devineni; Rahel Ghebre; Goutam Ghosh; Hemant Joshi; Yawu Jing; Alexander M. Truskinovsky; Sundaram Ramakrishnan

Endostatin potentiates the antimitotic effects of paclitaxel (taxol) on endothelial cells (ECs). P125A-endostatin and taxol-treated ECs showed multipolar spindles and nuclear lobulation, leading to mitotic catastrophe and cell death. Induction of nuclear abnormalities was found to be dependent on β-catenin levels as wnt-mediated overexpression of β-catenin reversed the changes in nuclear morphology. These results prompted us to investigate whether antiangiogenic gene therapy and paclitaxel chemotherapy can synergistically inhibit angiogenesis and tumor growth. We first determined the effect of combination treatment in a transgenic mouse model of breast cancer. Intramuscular injection of recombinant adeno-associated virus type-2 virus induced sustained expression of P125A-endostatin. In vivo studies showed that combination therapy inhibited mammary cancer growth, delayed the onset of multifocal mammary adenocarcinomas, decreased tumor angiogenesis and increased survival in treated mice. In a second model, female athymic mice were orthotopically transplanted with a metastatic human breast cancer cell line. Antiangiogenic gene therapy in combination with paclitaxel inhibited tumor angiogenesis and lung/lymph-node metastasis in this model. These studies demonstrate cooperation between endostatin gene therapy and chemotherapy to inhibit tumor initiation, growth and metastasis.


Cancer Research | 2011

Abstract 3980: Hypoxia-induced microRNA-424 targets CUL2 to stabilize HIF-α isoforms and promotes angiogenesis

Indira V. Subramanian; Goutam Ghosh; Julia Nguyen; Xiaoxiao Zhang; Hemant Joshi; Yan Zeng; Sundaram Ramakrishnan

Chronic hypoxia, a hallmark of many tumors, is associated with angiogenesis and tumor progression. Adaptive changes to oxygen availability are critical for cell survival and homeostasis. Cells respond to hypoxia by modulating oxygen-sensing transducers that stabilize the transcription factor hypoxia-inducible factor 1α (HIF-1α), which transactivates genes governing angiogenesis and metabolic pathways. Oxygen-dependent changes in HIF-1α levels are regulated by proline hydroxylation and proteasomal degradation. Here we provide evidence for a novel mechanism regulating HIF-1α and HIF-2α levels in endothelial cells during hypoxia. Hypoxia differentially increased microRNA-424 (miR-424) levels in microvascular EC, HUVEC and EC progenitor cells purified from the blood. miR-424 targeted cullin 2 (CUL2), a scaffolding protein critical to the assembly of the ubiquitin ligase system, thereby stabilizing HIF-α isoforms. Hypoxia-induced miR-424 in EC was regulated by PU.1-dependent transactivation. PU.1 expression was in turn controlled by RUNX-1 and C/EBPα. Knocking down either C/EBPα or RUNX-1 or PU.1 attenuated hypoxia-driven expression of miR-424 and nuclear translocation of HIF-1α. Furthermore, miR-424 promoted angiogenesis in vitro and in mice, which was blocked by specific morpholinos. These results suggest that miR-424 plays an important physiological role in angiogenesis. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3980. doi:10.1158/1538-7445.AM2011-3980

Collaboration


Dive into the Indira V. Subramanian's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Goutam Ghosh

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Hemant Joshi

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Zeng

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Yawu Jing

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huarui Lu

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Kailang Wu

University of Minnesota

View shared research outputs
Researchain Logo
Decentralizing Knowledge