Network


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

Hotspot


Dive into the research topics where Prahlad T. Ram is active.

Publication


Featured researches published by Prahlad T. Ram.


Nature Reviews Drug Discovery | 2005

Exploiting the PI3K/AKT Pathway for Cancer Drug Discovery

Bryan T. Hennessy; Debra L. Smith; Prahlad T. Ram; Yiling Lu; Gordon B. Mills

Evolving studies with several different targeted therapeutic agents are demonstrating that patients with genomic alterations of the target, including amplification, translocation and mutation, are more likely to respond to the therapy. Recent studies indicate that numerous components of the phosphatidylinositol-3-kinase (PI3K)/AKT pathway are targeted by amplification, mutation and translocation more frequently than any other pathway in cancer patients, with resultant activation of the pathway. This warrants exploiting the PI3K/AKT pathway for cancer drug discovery.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes

Joseph H. Taube; Jason I. Herschkowitz; Kakajan Komurov; Alicia Y. Zhou; Supriya Gupta; Jing Yang; Kimberly A. Hartwell; Tamer T. Onder; Piyush B. Gupta; Kurt W. Evans; Brett G. Hollier; Prahlad T. Ram; Eric S. Lander; Jeffrey M. Rosen; Robert A. Weinberg; Sendurai A. Mani

The epithelial-to-mesenchymal transition (EMT) produces cancer cells that are invasive, migratory, and exhibit stem cell characteristics, hallmarks of cells that have the potential to generate metastases. Inducers of the EMT include several transcription factors (TFs), such as Goosecoid, Snail, and Twist, as well as the secreted TGF-β1. Each of these factors is capable, on its own, of inducing an EMT in the human mammary epithelial (HMLE) cell line. However, the interactions between these regulators are poorly understood. Overexpression of each of the above EMT inducers up-regulates a subset of other EMT-inducing TFs, with Twist, Zeb1, Zeb2, TGF-β1, and FOXC2 being commonly induced. Up-regulation of Slug and FOXC2 by either Snail or Twist does not depend on TGF-β1 signaling. Gene expression signatures (GESs) derived by overexpressing EMT-inducing TFs reveal that the Twist GES and Snail GES are the most similar, although the Goosecoid GES is the least similar to the others. An EMT core signature was derived from the changes in gene expression shared by up-regulation of Gsc, Snail, Twist, and TGF-β1 and by down-regulation of E-cadherin, loss of which can also trigger an EMT in certain cell types. The EMT core signature associates closely with the claudin-low and metaplastic breast cancer subtypes and correlates negatively with pathological complete response. Additionally, the expression level of FOXC1, another EMT inducer, correlates strongly with poor survival of breast cancer patients.


Cancer Research | 2010

Basal and Treatment-Induced Activation of AKT Mediates Resistance to Cell Death by AZD6244 (ARRY-142886) in Braf-Mutant Human Cutaneous Melanoma Cells

Y.N. Vashisht Gopal; Wanleng Deng; Scott E. Woodman; Kakajan Komurov; Prahlad T. Ram; Paul D. Smith; Michael A. Davies

The majority of melanomas show constitutive activation of the RAS-RAF-MAP/ERK kinase (MEK)-mitogen-activated protein kinase (MAPK) pathway. AZD6244 is a selective MEK1/2 inhibitor that markedly reduces tumor P-MAPK levels, but it produces few clinical responses in melanoma patients. An improved understanding of the determinants of resistance to AZD6244 may lead to improved patient selection and effective combinatorial approaches. The effects of AZD6244 on cell growth and survival were tested in a total of 14 Braf-mutant and 3 wild-type human cutaneous melanoma cell lines. Quantitative assessment of phospho-protein levels in the Braf-mutant cell lines by reverse phase protein array (RPPA) analysis showed no significant association between P-MEK or P-MAPK levels and AZD6244 sensitivity, but activation-specific markers in the phosphoinositide 3-kinase (PI3K)-AKT pathway correlated with resistance. We also identified resistant cell lines without basal activation of the PI3K-AKT pathway. RPPA characterization of the time-dependent changes in signaling pathways revealed that AZD6244 produced durable and potent inhibition of P-MAPK in sensitive and resistant Braf-mutant cell lines, but several resistant lines showed AZD6244-induced activation of AKT. In contrast, sensitive cell lines showed AZD6244 treatment-induced upregulation of PTEN protein and mRNA expression. Inhibition of AKT, TORC1/2, or insulin-like growth factor I receptor blocked AZD6244-induced activation of AKT and resulted in synergistic cell killing with AZD6244. These findings identify basal and treatment-induced regulation of the PI3K-AKT pathway as a critical regulator of AZD6244 sensitivity in Braf-mutant cutaneous melanoma cells and the novel regulation of PTEN expression by AZD6244 in sensitive cells, and suggest new combinatorial approaches for patients.


Nature Communications | 2013

Tumour angiogenesis regulation by the miR-200 family

Chad V. Pecot; Rajesha Rupaimoole; Da Yang; Rehan Akbani; Cristina Ivan; Chunhua Lu; Sherry Y. Wu; Hee Dong Han; Maitri Y. Shah; Cristian Rodriguez-Aguayo; Justin Bottsford-Miller; Yuexin Liu; Sang Bae Kim; Anna K. Unruh; Vianey Gonzalez-Villasana; Li Huang; Behrouz Zand; Myrthala Moreno-Smith; Lingegowda S. Mangala; Morgan Taylor; Heather J. Dalton; Vasudha Sehgal; Yunfei Wen; Yu Kang; Keith A. Baggerly; Ju Seog Lee; Prahlad T. Ram; Murali Ravoori; Vikas Kundra; Xinna Zhang

The miR-200 family is well known to inhibit the epithelial-mesenchymal transition, suggesting it may therapeutically inhibit metastatic biology. However, conflicting reports regarding the role of miR-200 in suppressing or promoting metastasis in different cancer types have left unanswered questions. Here we demonstrate a difference in clinical outcome based on miR-200s role in blocking tumour angiogenesis. We demonstrate that miR-200 inhibits angiogenesis through direct and indirect mechanisms by targeting interleukin-8 and CXCL1 secreted by the tumour endothelial and cancer cells. Using several experimental models, we demonstrate the therapeutic potential of miR-200 delivery in ovarian, lung, renal and basal-like breast cancers by inhibiting angiogenesis. Delivery of miR-200 members into the tumour endothelium resulted in marked reductions in metastasis and angiogenesis, and induced vascular normalization. The role of miR-200 in blocking cancer angiogenesis in a cancer-dependent context defines its utility as a potential therapeutic agent.


Nature Communications | 2014

A pan-cancer proteomic perspective on The Cancer Genome Atlas

Rehan Akbani; Patrick Kwok Shing Ng; Henrica Maria Johanna Werner; Maria Shahmoradgoli; Fan Zhang; Zhenlin Ju; Wenbin Liu; Ji Yeon Yang; Kosuke Yoshihara; Jun Li; Shiyun Ling; Elena G. Seviour; Prahlad T. Ram; John D. Minna; Lixia Diao; Pan Tong; John V. Heymach; Steven M. Hill; Frank Dondelinger; Nicolas Städler; Lauren Averett Byers; Funda Meric-Bernstam; John N. Weinstein; Bradley M. Broom; Roeland Verhaak; Han Liang; Sach Mukherjee; Yiling Lu; Gordon B. Mills

Protein levels and function are poorly predicted by genomic and transcriptomic analysis of patient tumors. Therefore, direct study of the functional proteome has the potential to provide a wealth of information that complements and extends genomic, epigenomic and transcriptomic analysis in The Cancer Genome Atlas (TCGA) projects. Here we use reverse-phase protein arrays to analyze 3,467 patient samples from 11 TCGA “Pan-Cancer” diseases, using 181 high-quality antibodies that target 128 total proteins and 53 post-translationally modified proteins. The resultant proteomic data is integrated with genomic and transcriptomic analyses of the same samples to identify commonalities, differences, emergent pathways and network biology within and across tumor lineages. In addition, tissue-specific signals are reduced computationally to enhance biomarker and target discovery spanning multiple tumor lineages. This integrative analysis, with an emphasis on pathways and potentially actionable proteins, provides a framework for determining the prognostic, predictive and therapeutic relevance of the functional proteome.


Cancer Research | 2010

Identification of Optimal Drug Combinations Targeting Cellular Networks: Integrating Phospho-Proteomics and Computational Network Analysis

Sergio Iadevaia; Yiling Lu; Fabiana C. Morales; Gordon B. Mills; Prahlad T. Ram

Targeted therapeutics hold tremendous promise in inhibiting cancer cell proliferation. However, targeting proteins individually can be compensated for by bypass mechanisms and activation of regulatory loops. Designing optimal therapeutic combinations must therefore take into consideration the complex dynamic networks in the cell. In this study, we analyzed the insulin-like growth factor (IGF-1) signaling network in the MDA-MB231 breast cancer cell line. We used reverse-phase protein array to measure the transient changes in the phosphorylation of proteins after IGF-1 stimulation. We developed a computational procedure that integrated mass action modeling with particle swarm optimization to train the model against the experimental data and infer the unknown model parameters. The trained model was used to predict how targeting individual signaling proteins altered the rest of the network and identify drug combinations that minimally increased phosphorylation of other proteins elsewhere in the network. Experimental testing of the modeling predictions showed that optimal drug combinations inhibited cell signaling and proliferation, whereas nonoptimal combination of inhibitors increased phosphorylation of nontargeted proteins and rescued cells from cell death. The integrative approach described here is useful for generating experimental intervention strategies that could optimize drug combinations and discover novel pharmacologic targets for cancer therapy.


eLife | 2016

Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism

Hongyun Zhao; Lifeng Yang; Joelle Baddour; Abhinav Achreja; Vincent Bernard; Tyler Moss; Juan C. Marini; Thavisha Tudawe; Elena G. Seviour; F. Anthony San Lucas; Hector Alvarez; Sonal Gupta; Sourindra Maiti; Laurence J.N. Cooper; Donna M. Peehl; Prahlad T. Ram; Anirban Maitra; Deepak Nagrath

Cancer-associated fibroblasts (CAFs) are a major cellular component of tumor microenvironment in most solid cancers. Altered cellular metabolism is a hallmark of cancer, and much of the published literature has focused on neoplastic cell-autonomous processes for these adaptations. We demonstrate that exosomes secreted by patient-derived CAFs can strikingly reprogram the metabolic machinery following their uptake by cancer cells. We find that CAF-derived exosomes (CDEs) inhibit mitochondrial oxidative phosphorylation, thereby increasing glycolysis and glutamine-dependent reductive carboxylation in cancer cells. Through 13C-labeled isotope labeling experiments we elucidate that exosomes supply amino acids to nutrient-deprived cancer cells in a mechanism similar to macropinocytosis, albeit without the previously described dependence on oncogenic-Kras signaling. Using intra-exosomal metabolomics, we provide compelling evidence that CDEs contain intact metabolites, including amino acids, lipids, and TCA-cycle intermediates that are avidly utilized by cancer cells for central carbon metabolism and promoting tumor growth under nutrient deprivation or nutrient stressed conditions. DOI: http://dx.doi.org/10.7554/eLife.10250.001


Molecular Systems Biology | 2014

Metabolic shifts toward glutamine regulate tumor growth, invasion and bioenergetics in ovarian cancer

Lifeng Yang; Tyler Moss; Lingegowda S. Mangala; Juan C. Marini; Hongyun Zhao; Stephen Wahlig; Guillermo N. Armaiz-Pena; Dahai Jiang; Abhinav Achreja; Julia Win; Rajesha Roopaimoole; Cristian Rodriguez-Aguayo; Imelda Mercado-Uribe; Gabriel Lopez-Berestein; Jinsong Liu; Takashi Tsukamoto; Anil K. Sood; Prahlad T. Ram; Deepak Nagrath

Glutamine can play a critical role in cellular growth in multiple cancers. Glutamine‐addicted cancer cells are dependent on glutamine for viability, and their metabolism is reprogrammed for glutamine utilization through the tricarboxylic acid (TCA) cycle. Here, we have uncovered a missing link between cancer invasiveness and glutamine dependence. Using isotope tracer and bioenergetic analysis, we found that low‐invasive ovarian cancer (OVCA) cells are glutamine independent, whereas high‐invasive OVCA cells are markedly glutamine dependent. Consistent with our findings, OVCA patients’ microarray data suggest that glutaminolysis correlates with poor survival. Notably, the ratio of gene expression associated with glutamine anabolism versus catabolism has emerged as a novel biomarker for patient prognosis. Significantly, we found that glutamine regulates the activation of STAT3, a mediator of signaling pathways which regulates cancer hallmarks in invasive OVCA cells. Our findings suggest that a combined approach of targeting high‐invasive OVCA cells by blocking glutamines entry into the TCA cycle, along with targeting low‐invasive OVCA cells by inhibiting glutamine synthesis and STAT3 may lead to potential therapeutic approaches for treating OVCAs.


Oncogene | 2001

G protein coupled receptor signaling through the Src and Stat3 pathway : role in proliferation and transformation

Prahlad T. Ram; Ravi Iyengar

Extracellular signals when routed through signaling pathways that use heterotrimeric G proteins can engage multiple signaling pathways leading to diverse biological consequences. One locus at which signal sorting occurs is at the level of G proteins. G protein α-subunits appear to be capable of interacting with different effectors leading to engagement of distinct signaling pathways. Regulation of different pathways in turn leads to different biological outcomes. The process of neoplastic transformation is controlled to a large extent through the activation and inhibition of signaling pathways. Signaling pathways such as the Ras-MAPK, v-Src-Stat3 pathways are activated in the process of transformation. Expression of activated Gα subunits have been shown to cause transformation of cells. While activation of the MAPK 1,2 pathway by various Gα subunits has been reported for several years, recent studies show the activation and involvement of Src and Stat3 pathways in Gαo and Gαi mediated transformation of cells. Recent studies also suggest that both Gαi and Gαs may be able to interact with and activate Src. The activation of Src and Stat3 by G proteins has also been demonstrated by ligand-induced activation of G protein receptors. So increasingly it is becoming clear that the Src and Stat3 pathways are potential effectors for G proteins and that they may play a role in G protein function.


Cancer Cell | 2014

Hematogenous Metastasis of Ovarian Cancer: Rethinking Mode of Spread

Sunila Pradeep; Seung W. Kim; Sherry Y. Wu; Masato Nishimura; Pradeep Chaluvally-Raghavan; Takahito Miyake; Chad V. Pecot; Sun Jin Kim; Hyun Jin Choi; Farideh Z. Bischoff; Julie Ann Mayer; Li Huang; Alpa M. Nick; Carolyn S. Hall; Cristian Rodriguez-Aguayo; Behrouz Zand; Heather J. Dalton; Thiruvengadam Arumugam; Ho Jeong Lee; Hee Dong Han; Min Soon Cho; Rajesha Rupaimoole; Lingegowda S. Mangala; Vasudha Sehgal; Sang Cheul Oh; Jinsong Liu; Ju Seog Lee; Robert L. Coleman; Prahlad T. Ram; Gabriel Lopez-Berestein

Ovarian cancer has a clear predilection for metastasis to the omentum, but the underlying mechanisms involved in ovarian cancer spread are not well understood. Here, we used a parabiosis model that demonstrates preferential hematogenous metastasis of ovarian cancer to the omentum. Our studies revealed that the ErbB3-neuregulin 1 (NRG1) axis is a dominant pathway responsible for hematogenous omental metastasis. Elevated levels of ErbB3 in ovarian cancer cells and NRG1 in the omentum allowed for tumor cell localization and growth in the omentum. Depletion of ErbB3 in ovarian cancer impaired omental metastasis. Our results highlight hematogenous metastasis as an important mode of ovarian cancer metastasis. These findings have implications for designing alternative strategies aimed at preventing and treating ovarian cancer metastasis.

Collaboration


Dive into the Prahlad T. Ram's collaboration.

Top Co-Authors

Avatar

Anil K. Sood

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gordon B. Mills

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Vasudha Sehgal

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cristian Rodriguez-Aguayo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gabriel Lopez-Berestein

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tyler Moss

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rajesha Rupaimoole

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sunila Pradeep

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sherry Y. Wu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yiling Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge