Aug
12
Mon
2013
Plenary Address: A novel strategy for targeting metalloproteinases in cancer @ Acharya Hall
Aug 12 @ 1:30 pm – 2:00 pm

gillianGillian Murphy, Ph.D.
Professor, Department of Oncology, University of Cambridge, UK


A novel strategy for targeting metalloproteinases in cancer

Epithelial tumours evolve in a multi-step manner, involving both inflammatory and mesenchymal cells. Although intrinsic factors drive malignant progression, the influence of the micro-environment of neoplastic cells is a major feature of tumorigenesis. Extracellular proteinases, notably the metalloproteinases, are key players in the regulation of this cellular environment, acting as major effectors of both cell-cell and cell-extracellular matrix (ECM) interactions. They are involved in modifying ECM integrity, growth factor availability and the function of cell surface signalling systems, with consequent effects on cellular differentiation, proliferation and apoptosis.This has made metalloproteinases important targets for therapeutic interventions in cancer and small molecule inhibitors focussed on chelation of the active site zinc and binding within the immediate active site pocket were developed.  These were not successful in early clinical trials due to the relative lack of specificity and precise knowledge of the target proteinase(s) in specific cancers. We can now appreciate that it is essential that we understand the relative roles of the different enzymes (of which there are over 60) in terms of their pro and anti tumour activity and their precise sites of expression The next generations of metalloproteinase inhibitors need the added specificity that might be gained from an understanding of the structure of individual active sites and the role of extra catalytic domains in substrate binding and other aspects of their biology. We have prepared scFv antibodies to the extra catalytic domains of two membrane metalloproteinases, MMP-14 and ADAM17, that play key roles in the tumour microenvironment. Our rationale and experiences with these agents will be presented in more detail.

Gillian

Invited Talk: Identification of Potential Early Diagnostic Biomarkers for Gliomas and Various Infectious Diseases using Proteomic Technologies @ Acharya Hall
Aug 12 @ 2:35 pm – 2:56 pm

SanjeevaSanjeeva Srivastava, Ph.D.
Assistant Professor, Proteomics Lab, IIT-Bombay, India


Identification of Potential Early Diagnostic Biomarkers for Gliomas and Various Infectious Diseases using Proteomic Technologies 

The spectacular advancements achieved in the field of proteomics research during the last decade have propelled the growth of proteomics for clinical research. Recently, comprehensive proteomic analyses of different biological samples such as serum or plasma, tissue, CSF, urine, saliva etc. have attracted considerable attention for the identification of protein biomarkers as early detection surrogates for diseases (Ray et al., 2011). Biomarkers are biomolecules that can be used for early disease detection, differentiation between closely related diseases with similar clinical manifestations as well as aid in scrutinizing disease progression. Our research group is performing in-depth analysis of alteration in human proteome in different types of brain tumors and various pathogenic infections to obtain mechanistic insight about the disease pathogenesis and host immune responses, and identification of surrogate protein markers for these fatal human diseases.

Applying 2D-DIGE in combination with MALDI-TOF/TOF MS we have analyzed the serum and tissue proteome profiles of glioblastoma multiforme; the most common and lethal adult malignant brain tumor (Gollapalli et al., 2012) (Figure 1). Results obtained were validated by employing different immunoassay-based approaches. In serum proteomic analysis we have identified some interesting proteins like haptoglobin, ceruloplasmin, vitamin-D binding protein etc. Moreover, proteomic analysis of different grades (grade-I to IV) of gliomas and normal brain tissue was performed and differential expressions of quite a few proteins such as SIRT2, GFAP, SOD, CDC42 have been identified, which have significant correlation with the tumor growth. While proteomic analysis of cerebrospinal fluid from low grade (grade I & II) vs. high grade (grade III & IV) gliomas revealed modulation of CSF levels of apolipoprotein E, dickkopf related protein 3, vitamin D binding protein and albumin in high grade gliomas. The prospective candidates identified in our studies provide a mechanistic insight of glioma pathogenesis and identification of potential biomarkers. We are also studying the role of JAK/STAT interactome and therapeutic potential of STAT3 inhibitors in gliomas using proteomics approach. Several candidates of the JAK/STAT interactome were identified with altered expression and a significant correlation was observed between STAT3 and PDK1 transcript expression level.

We have also investigated the changes in human serum proteome in different infectious diseases including falciparum and vivax malaria (Ray et al., 2012a; Ray et al., 2012b), dengue (Ray et al., 2012c) and leptospirosis (Srivastava et al., 2012). Although, quite a few serum proteins were found to be commonly altered in different infectious diseases and might be a consequence of inflammation mediated acute phase response signaling, uniquely modulated candidates were identified in each pathogenic infection indicating the some inimitable responses. Further, a panel of identified proteins consists of six candidates; serum amyloid A, hemopexin, apolipoprotein E, haptoglobin, retinol-binding protein and apolipoprotein A-I was used to build statistical sample class prediction models employing PLSDA and other classification methods to predict the clinical phenotypic classes and 91.37% overall prediction accuracy was achieved (Figure 2). ROC curve analysis was carried out to evaluate the individual performance of classifier proteins. The excellent discrimination among the different disease groups on the basis of differentially expressed proteins demonstrates the potential diagnostic implications of this analytical approach.

Keywords: Diagnostic biomarkers, Gliomas, Infectious Diseases, Proteomics, Serum proteome

Acknowledgments: This disease biomarker discovery research was supported by Department of Biotechnology, India grant (No. BT/PR14359/MED/30/916/2010), Board of Research in Nuclear Sciences (BRNS) DAE young scientist award (2009/20/37/4/BRNS) and a startup grant 09IRCC007 from the IIT Bombay. The active support from Advanced Center for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Hospital (TMH), and Seth GS Medical College and KEM Hospital Mumbai, India in clinical sample collection process is gratefully acknowledged.

References :

  1. Ray S, Reddy PJ, Jain R, Gollapalli K. Moiyadi A, Srivastava S. Proteomic technologies for the identification of disease biomarkers in serum: advances and challenges ahead. Proteomics 11: 2139-61, 2011.
  2. Gollapalli K, Ray S, Srivastava R, Renu D, Singh P, Dhali S, Dikshit JB, Srikanth R, Moiyadi A, Srivastava S. Investigation of serum proteome alterations in human glioblastoma multiforme. Proteomics 12(14): 2378-90, 2012.
  3. Ray S, Renu D, Srivastava R, Gollapalli K, Taur S, Jhaveri T, Dhali S, Chennareddy S, Potla A, Dikshit JB, Srikanth R, Gogtay N, Thatte U, Patankar S, Srivastava S. Proteomic investigation of falciparum and vivax malaria for identification of surrogate protein markers. PLoS One 7(8): e41751, 2012a.
  4. Ray S, Kamath KS, Srivastava R, Raghu D, Gollapalli K, Jain R, Gupta SV, Ray S, Taur S, Dhali S, Gogtay N, Thatte U, Srikanth R, Patankar S, Srivastava S. Serum proteome analysis of vivax malaria: An insight into the disease pathogenesis and host immune response. J Proteomics 75(10): 3063-80, 2012b.
  5. Srivastava R, Ray S, Vaibhav V, Gollapalli K, Jhaveri T, Taur S, Dhali S, Gogtay N, Thatte U, Srikanth R, Srivastava S. Serum profiling of leptospirosis patients to investigate proteomic alterations. J Proteomics 76: 56-68, 2012.
  6. Ray S, Srivastava R, Tripathi K, Vaibhav V, Srivastava S. Serum proteome changes in dengue virus-infected patients from a dengue-endemic area of India: towards new molecular targets? OMICS 16(10): 527-36, 2012c.

* Correspondence: Dr. Sanjeeva Srivastava, Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India: E-mail: sanjeeva@iitb.ac.in; Phone: +91-22-2576-7779, Fax: +91-22-2572-3480

Figure 1 (a) Differentially expressed proteins in GBM identified using 2D-DIGE. Representative 2D- DIGE image to compare serum proteome of HC and GBM patients. GBM and HC samples were labeled with Cy3 and Cy5 respectively, while the protein reference pool (internal standard) was labeled with Cy2. Graphical and 3D fluorescence intensity representations of few selected statistically significant (p < 0.05) differentially expressed proteins in GBM patients identified in biological variation analysis (BVA) using DeCyder 2D software. (b) Involvement of different essential physiological pathways with differentially expressed proteins in GBM. Members of multiple essential physiological processes including cell growth and proliferation, vitamin D metabolism, lipoprotein metabolism and transport, oxidative stress regulation, complement cascade, and platelet activation found to be modulated in the GBM patients (Gollapalli et al., Proteomics 2012).
Figure 1 (a) Differentially expressed proteins in GBM identified using 2D-DIGE. Representative 2D- DIGE image to compare serum proteome of HC and GBM patients. GBM and HC samples were labeled with Cy3 and Cy5 respectively, while the protein reference pool (internal standard) was labeled with Cy2. Graphical and 3D fluorescence intensity representations of few selected statistically significant (p < 0.05) differentially expressed proteins in GBM patients identified in biological variation analysis (BVA) using DeCyder 2D software. (b) Involvement of different essential physiological pathways with differentially expressed proteins in GBM. Members of multiple essential physiological processes including cell growth and proliferation, vitamin D metabolism, lipoprotein metabolism and transport, oxidative stress regulation, complement cascade, and platelet activation found to be modulated in the GBM patients (Gollapalli et al., Proteomics 2012).
Figure 2 (a) Western blot analysis of haptoglobin (HP), serum amyloid A (SAA), and clusterin (CLU) from serum samples of healthy control (HC) [n = 12], falciparum malaria (FM) [n = 12], vivax malaria (VM) [n = 12], Leptospirosis (Lep) [n = 6], dengue fever [DF] [n = 6] and non infectious disease control (NIDC:GBM) [n = 12]. Representative blots of the target proteins are depicted along with their respective relative abundance volumes (volume X 104). All the data are represented as mean ± SE. (b) Discrimination of malaria from dengue, leptospirosis and GBM using PLS-DA analysis. PLS-DA scores Plot for FM (blue spheres, n = 8), VM (green spheres, n = 8), DF (red spheres, n = 6), Lep (grey spheres, n = 6) and GBM (brown spheres, n = 8) samples based on 6 differentially expressed proteins (serum amyloid A, hemopexin, apolipoprotein E, haptoglobin, retinol-binding protein and apolipoprotein A-I) identified using DIGE. The axes of the plot indicate PLSDA latent variables t0-t2.
Figure 2 (a) Western blot analysis of haptoglobin (HP), serum amyloid A (SAA), and clusterin (CLU) from serum samples of healthy control (HC) [n = 12], falciparum malaria (FM) [n = 12], vivax malaria (VM) [n = 12], Leptospirosis (Lep) [n = 6], dengue fever [DF] [n = 6] and non infectious disease control (NIDC:GBM) [n = 12]. Representative blots of the target proteins are depicted along with their respective relative abundance volumes (volume X 104). All the data are represented as mean ± SE. (b) Discrimination of malaria from dengue, leptospirosis and GBM using PLS-DA analysis. PLS-DA scores Plot for FM (blue spheres, n = 8), VM (green spheres, n = 8), DF (red spheres, n = 6), Lep (grey spheres, n = 6) and GBM (brown spheres, n = 8) samples based on 6 differentially expressed proteins (serum amyloid A, hemopexin, apolipoprotein E, haptoglobin, retinol-binding protein and apolipoprotein A-I) identified using DIGE. The axes of the plot indicate PLSDA latent variables t0-t2.

 

Sanjeeva (1) Sanjeeva (2)

Aug
13
Tue
2013
Plenary Talk: Biosensor and Single Cell Manipulation using Nanopipettes @ Amriteshwari Hall
Aug 13 @ 10:06 am – 10:49 am

NaderNader Pourmand, Ph.D.
Director, UCSC Genome Technology Center,University of California, Santa Cruz


Biosensor and Single Cell Manipulation using Nanopipettes

Approaching sub-cellular biological problems from an engineering perspective begs for the incorporation of electronic readouts. With their high sensitivity and low invasiveness, nanotechnology-based tools hold great promise for biochemical sensing and single-cell manipulation. During my talk I will discuss the incorporation of electrical measurements into nanopipette technology and present results showing the rapid and reversible response of these subcellular sensors  to different analytes such as antigens, ions and carbohydrates. In addition, I will present the development of a single-cell manipulation platform that uses a nanopipette in a scanning ion-conductive microscopy technique. We use this newly developed technology to position the nanopipette with nanoscale precision, and to inject and/or aspirate a minute amount of material to and from individual cells or organelle without comprising cell viability. Furthermore, if time permits, I will show our strategy for a new, single-cell DNA/ RNA sequencing technology that will potentially use nanopipette technology to analyze the minute amount of aspirated cellular material.

Invited Talk: “Inside-out” NF-kappa B signaling in cancer and other pathologies @ Acharya Hall
Aug 13 @ 11:25 am – 11:40 am

ShigekiShigeki Miyamoto, Ph.D.
Professor, McArdle Laboratory for Cancer Research – UW Carbone Cancer Center
Department of Oncology, School of Medicine and Public Health
University of Wisconsin-Madison


“Inside-out” NF-κB signaling in cancer and other pathologies

The NF-κB/Rel family of transcription factors contributes to critical cellular processes, including immune, inflammatory and cell survival responses. As such, NF-κB is implicated in immunity-related diseases, as well as multiple types of human malignancies. Indeed, genetic alterations in the NF-κB signaling pathway are frequently observed in multiple human malignancies. NF-κB is normally kept inactive in the cytoplasm by inhibitor proteins. Extracellular ligands can induce the release of NF-κB from the inhibitors to allow its migration into the nucleus to regulate a variety of target genes.  NF-κB activation is also induced in response to multiple stress conditions, including those induced by DNA-damaging anticancer agents. Although precise mechanisms are still unclear, research from our group has revealed a unique nuclear-to-cytoplasmic signaling pathway. In collaboration with bioengineers, clinicians and pharmaceutical industry, our lab has developed new methods to analyze primary cancer patient samples and identified several compounds with different mechanisms that mitigate this cell survival pathway.  Further contributions from other labs have also revealed additional mechanisms and molecular players in this “inside-out” signaling pathway and expanded its role in other physiological and pathological processes, including B cell development, premature aging and therapy resistance of certain cancers. Our own new findings, along with these recent developments in the field, will be highlighted.

Shigeki

Invited Talk: Nanoscale Simulations – Tackling Form and Formulation Challenges in Drug Development and Drug Delivery @ Sathyam Hall
Aug 13 @ 2:15 pm – 2:40 pm

lalithaLalitha Subramanian, Ph.D.
Chief Scientific Officer & VP, Services at Scienomics, USA


Nanoscale Simulations – Tackling Form and Formulation Challenges in Drug Development and Drug Delivery

Lalitha Subramanian, Dora Spyriouni, Andreas Bick, Sabine Schweizer, and Xenophon Krokidis Scienomics

The discovery of a compound which is potent in activity against a target is a major milestone in Pharmaceutical and Biotech industry. However, a potent compound is only effective as a therapeutic agent when it can be administered such that the optimal quantity is transported to the site of action at an optimal rate. The active pharmaceutical ingredient (API) has to be tested for its physicochemical properties before the appropriate dosage form and formulation can be designed. Some of the commonly evaluated parameters are crystal forms and polymorphs, solubility, dissolution behavior, stability, partition coefficient, water sorption behavior, surface properties, particle size and shape, etc. Pharmaceutical development teams face the challenge of quickly and efficiently determining a number of properties with small quantities of the expensive candidate compounds. Recently the trend has been to screen these properties as early as possible and often the candidate compounds are not available in sufficient quantities. Increasingly, these teams are leveraging nanoscale simulations similar to those employed by drug discovery teams for several decades. Nanoscale simulations are used to predict the behavior using very little experimental data and only if this is promising further experiments are done. Another aspect where nanoscale simulations are being used in drug development and drug delivery is to get insights into the behavior of the system so that process failures can be remediated and formulation performance can be improved. Thus, the predictive screening and the in-depth understanding leads to experimental efficiency resulting in far-reaching business impacts.

With specific examples, this talk will focus on the different types of nanoscale simulations used to predict properties of the API in excipients and also provide insight into system behavior as a function of shelf life, temperature, mechanical stress, etc.

Invited Talk: Probing Estrogen Receptor – Tumor Suppressor p53 Interaction in Cancer: From Basic Research to Clinical Trial @ Acharya Hall
Aug 13 @ 3:26 pm – 3:57 pm

gokuldasGokul Das, Ph.D.
Co-Director, Breast Disease Site Research Group, Roswell Park Cancer Institute, Buffalo, NY


Probing Estrogen Receptor−Tumor Suppressor p53 Interaction in Cancer: From Basic Research to Clinical Trial

Tumor suppressor p53 and estrogen receptor have opposite roles in the onset and progression of breast cancer. p53 responds to a variety of cellular of stresses by restricting the proliferation and survival of abnormal cells. Estrogen receptor plays an important role in normal mammary gland development and the preservation of adult mammary gland function; however, when deregulated it becomes abnormally pro-proliferative and greatly contributes to breast tumorigenesis. The biological actions of estrogens are mediated by two genetically distinct estrogen receptors (ERs): ER alpha and ER beta. In addition to its expression in several ER alpha-positive breast cancers and normal mammary cells, ER beta is usually present in ER alpha-negative cancers including triple-negative breast cancer. In spite of genetically being wild type, why p53 is functionally debilitated in breast cancer has remained unclear. Our recent finding that ER alpha binds directly to p53 and inhibits its function has provided a novel mechanism for inactivating genetically wild type p53 in human cancer. Using a combination of proliferation and apoptosis assays, RNAi technology, quantitative chromatin immunoprecipitation (qChIP), and quantitative real-time PCR (qRT-PCR), in situ proximity ligation assay (PLA), and protein expression analysis in patient tissue micro array (TMA), we have demonstrated binding of ER alpha to p53 and have delineated the domains on both the proteins necessary for the interaction. Importantly, ionizing radiation inhibits the ER-p53 interaction in vivo both in human cancer cells and human breast tumor xenografts in mice. In addition, antiestrogenstamoxifen and faslodex/fulvestrant (ICI 182780) disrupt the ER-p53 interaction and counteract the repressive effect of ER alpha on p53, whereas 17β-estradiol (E2) enhances the interaction. Intriguingly, E2 has diametrically opposite effects on corepressor recruitment to a p53-target gene promoter versus a prototypic ERE-containing promoter. Thus, we have uncovered a novel mechanism by which estrogen could be providing a strong proliferative advantage to cells by dual mechanisms: enhancing expression of ERE-containing pro-proliferative genes while at the same time inhibiting transcription of p53-dependent anti-proliferative genes. Consistently, ER alpha enhances cell cycle progression and inhibits apoptosis of breast cancer cells. Correlating with these observations, our retrospective clinical study shows that presence of wild type p53 in ER-positive breast tumors is associated with better response to tamoxifen therapy. These data suggest ER alpha-p53 interaction could be one of the mechanisms underlying resistance to tamoxifen therapy, a major clinical challenge encountered in breast cancer patients. We have launched a prospective clinical trial to analyze ER-p53 interaction in breast cancer patient tumors at Roswell Park Cancer Institute. Our more recent finding that ER beta has opposite functions depending on the mutational status of p53 in breast cancer cells is significant in understanding the hard-to-treat triple-negative breast cancer and in developing novel therapeutic strategies against it. Our integrated approach to analyze ER-p53 interaction at the basic, translational, and clinical research levels has major implications in the diagnosis, prognosis, and treatment of breast cancer.

 

Invited Talk: Cancer Stem Cells – Target Colon Cancer @ Acharya Hall
Aug 13 @ 4:25 pm – 5:04 pm

ShrikantShrikant Anant, Ph.D.
The Department of Molecular & Integrative Physiology, Kansas University Medical Center, USA


Cancer Stem Cells: Target Colon Cancers

Shrikant Anant, Deep Kwatra and Dharmalingam Subramaniam

Colon cancer is a leading cause of cancer related deaths in the US, and its rate is increasing at an alarming rate in lndia. Recent studies have suggested the drug resistance role for a mall number of cells within a tumor called cancer stem cells. We identified the colon cancer stem cell marker DCLK1, a member of the protein kinase superfamily and the doublecortin family. The protein encodes a Cterminal serinethreonine protein kinase domain, which shows substantial homology to Ca2calmodulindependent protein kinase. Our current studies have been to identify compounds that can either affect DCLK1 expression or inhibits its activity as a way to inhibit cancer stem cells. Honokiol is a biphenolic compound that has been used in the traditional Chinese Medicine for treating various ailments. In vitro kinase assays with recombinant DCLK1 demonstrated that honokiol inhibits its kinase activity in a dose dependent manner. We therefore determined the effect of honokiol on stem cells. One method to look at effects on stem cells is perform a spheroid assay, where spheroids formation is suggested to maintain stemlike characteristic of cancer cells. Honokiol significantly suppressed colonosphere formation of two colon cancer cell lines HCT116 and SW480. Flow cytometry studies confirmed that honokiol reduced the number of DCLK1cells. A critical signaling pathway known to modulate intestinal stem cell proliferation is the Hippo signaling pathway, and deregulation of the pathway leads to tumor development. DCLK1cells had high levels of YAP1, the nuclear target of Hippo signaling. We determined the effect of honokiol on components of the hipposignaling pathway. Honokiol reduced the phosphorylation of Mst1/2, Lats1/2 and YAP1. Furthermore, honokiol treatment resulted in downregulation of YAPTEAD complex protein TEAD-1. Ectopic expression of the TEAD-1 partially rescued the cells from honokiol mediated growth suppression. To determine the effect of honokiol on tumor growth in vivo, nude mice harboring HCT116 tumor xenografts in their flanks were administered the compound intraperitoneally every day for 21 days. Honokiol treatment significantly inhibited tumor xenograft growth. Western blot and immunohistochemistry analyses demonstrated significant inhibition in the expression of stem marker and Hippo signaling proteins in the honokioltreated xenograft tissues. Taken together, these data suggest that honokiol is a potent inhibitor of colon cancer that targets DCLK1 stem cells by inhibiting Hippo signaling pathway.

Delegate Talk: A Non-toxic Approach to Prevent Secondary Effects of Chemotherapies @ Acharya Hall
Aug 13 @ 6:26 pm – 6:38 pm
Delegate Talk: A Non-toxic Approach to Prevent Secondary Effects of Chemotherapies @ Acharya Hall | Vallikavu | Kerala | India

Tanu Sharma, Narendra Parihar, Someshwar Nath, Azad Singh, Savneet Kaur, Simendra Singh and Chandi Mandal.


In the recent years, there has been a surge in various forms of cancers especially in urban areas of India. Tumor recurrence and metastasis are the main causes of increased morbidity and mortality. In poor and developing countries like India chemotherapy is the choosen method of therapeutic intervention for different types of malignancies such as breast, prostate, pancreas, etc. Although general chemotherapy treatment is effective in controlling tumour growth but high doses of the treatment regimen often results in severe off-target toxicity to different organs including liver, kidney, heart, brain etc. (1) A large percentage of patients are unable to tolerate this toxicity and sometime it can also lead to life threatening complications. (2) Therefore, the primary objective of this study is to prevent this druginduced off-target toxicity. Omega 3 fatty acids (n-3FAs) such as DHA (docosahexaenoic acid) and EPA (eicosapentaenoic acid) that are active components of fish oil play active role in preventing cancer growth and its metastasis. (3–5) We have recently identified that omega 3-fatty acids prevent breast cancer bone metastasis by targeting cancer stem cell marker CD44 and osteoclastogenic factor CSF-1(4, 5). With this background, the present study addresses the role of n-3FAs in preventing chemotherapeutic drug-induced off-target toxicity.

Methods
The cat fishes were divided into four groups (each group contains 4 fishes) for each set of experiments. Three different chemotherapeutic drugs etoposide, doxorubicin, cisplatin at higher dosages (4, 2, 2 mg/kg body wt respectively) and fish oil (60mg/kg body wt, DHA: EPA; 2:3) were administered to cat fish (Mangur) by gavage. After 6 days, cat fishes were sacrificed and different organs were isolated. SGPT and alkaline phosphatase (ALP) activity assays were performed to examine liver function and superoxide dismutase activity (SOD) assay was conducted to evaluate reactive oxygen species (ROS) level.

Results
Severe skin damages were observed both in doxorubicin and cisplatintreated cat fishes as compared to the control fishes, indicating side effects of chemotherapeutic drugs treatment. High level of SGPT as well as ALP activity was observed in liver samples of etoposide and doxorubicin – treated fishes, suggesting chemotherapeutic drug-induced liver toxicity. Mechanistically, we found that doxorubicin treatment showed significant decrease of SOD activity in liver samples in comparison to the control fishes, suggesting that chemotherapeutic drugs lead to organ toxicity presumably by increasing ROS levels (6). However, treatment of the fishes with n-3FAs of the fish oil led to a dramatic reduction of chemotherapeutic drug-induced skin damages. Also, low levels of SGPT and ALP activities were observed in the fishes given etoposide in combination with n-3FAs as compared to the fishes given etoposide alone. Similarly, fish oil also gave protection against doxorubicin-induced liver dysfunction. Our data further showed that n-3FAs treatment significantly increased etoposide and doxorubicin-inhibited SOD activity.

Conclusions
The study for the first time reports that the use of n-3FAs leads to a dramatic reduction of chemotherapeutic drug-induced skin damages and mitigates chemotherapeutic drug-driven liver dysfunction presumably by reducing ROS level. This study suggests that n-3FAs may possibly be used in combination with chemotherapeutic drugs to treat different cancers to reduce chemotherapeutic drugs-associated systemic toxicity and to increase anticancer activity. Ongoing research study will further address the role of n-3FAs on other off-target toxic effects of chemotherapeutic drugs.

Delegate Talk: A proteomic approach to decipher the mechanism of action and molecular targets of curcumin in Bacillus subtilis @ Acharya Hall
Aug 13 @ 7:03 pm – 7:13 pm
Delegate Talk: A proteomic approach to decipher the mechanism of action and molecular targets of curcumin in Bacillus subtilis @ Acharya Hall | Vallikavu | Kerala | India

Jaipal Panga Reddy, Dulal Panda and Sanjeeva Srivastava


The rapid emergence of microbial resistance against the ever increasing number of infectious diseases has been a major hurdle in hunt for novel antibiotics and inventive targets required to combat these dreaded pathogens. Despite the discovery of synthetic and semi-synthetic drugs, infectious diseases remain a major cause of concern. The process of drug discovery started from natural products as an antibacterial drug, an old art which was widely adopted in ancient civilizations in India and China. Most of the existing antibiotics are derived from the backbone of natural compounds (Butler M S 2005). Drug discovery process from natural products to synthetic medicine has continued for thousands of years to battle with pathogenic organisms. Although synthetic drugs played a vital role as antimicrobial drugs during the last decade, the over-use of antibiotics has led to the unanticipated changes at the genomic level, resulting into emergence of antibiotic-resistant strains (Singh P et al. 2010). To combat drug-resistant strains there is a need to develop and characterize new drugs by screening natural and synthetic compounds. Curcumin is a well known natural product, with a potential to cure wide range of cancers. Apart from antitumor activity, it also has anti-inflammatory, anti-viral, anti-genotoxic, phototoxic and antimicrobial activities. The exact mechanism of action of curcumin is still obscure and further investigations are required to identify its molecular/cellular targets. Recently, it was observed that curcumin is able to perturb the FtsZ assembly dynamics and elongate the bacterial cell length by inhibiting bacterial cell division (Rai D et al. 2008).

In the present study, we aimed at deciphering the mechanism of action and molecular targets of curcumin in B. subtilis AH75 using classical two dimensional electrophoresis, 2D-difference gel electrophoresis (2DDIGE) in combination with MALDI-TOF/TOFMS. Comparative proteome analysis of control and IC50 curcumin treated B. subtilis has revealed differential expression of 48 proteins (p ≤ 0.05) in response to curcumin treatment. Further, in silico analysis has revealed the involvement of the differential expressed proteins in protein synthesis, transcription/nucleotide synthesis, stress regulation, central metabolism and amino acid metabolic process. Additionally, suppression in major central metabolic dehydrogenase such as glyceraldehyde-3-phosphate dehydrogenase 2, dihydrolipoyl dehydrogenase, malate dehydrogenase, pyruvate dehydrogenase E1 component subunit beta, 2-oxoglutarate dehydrogenase E1 component, ATP synthase epsilon chain, ATP synthase subunit beta and probable NADdependent malic enzyme 1 has been identified (Figure 1). Reduction in expression levels of major dehydrogenases indicates the disturbance in cell membrane integrity to transfer the electrons from NADH to molecular oxygen or maintain the PMF or maintain the membrane potential. Selected potential proteins obtained from proteomic analysis were validated with real-time expression analysis and metabolic assays such as resazurin assay for metabolic activity, CTC assay for respiratory activity and propidium iodide staining for membrane integrity. Findings from this study have revealed that curcumin has potential effects on multiple physiological pathways in bacteria and inhibition of the cell division machinery is one of the prime targets of this drug. Multiple proteins involved in cell division process such as GroEL, ClpC, MetK, Tuf and major dehydrogenases are significantly modulated as a consequence of the drug treatment.

Aug
14
Wed
2013
Invited Talk: A draft map of the human proteome @ Amriteshwari Hall
Aug 14 @ 10:42 am – 11:30 am

akhileshAkhilesh Pandey, Ph.D.
Professor, Johns Hopkins University School of Medicine, Baltimore, USA


A draft map of the human proteome

We have generated a draft map of the human proteome through a systematic and comprehensive analysis of normal human adult tissues, fetal tissues and hematopoietic cells as an India-US initiative. This unique dataset was generated from 30 histologically normal adult tissues, fetal tissues and purified primary hematopoietic cells that were analyzed at high resolution in the MS mode and by HCD fragmentation in the MS/MS mode on LTQ-Orbitrap Velos/Elite mass spectrometers. This dataset was searched against a 6-frame translation of the human genome and RNA-Seq transcripts in addition to standard protein databases. In addition to confirming a large majority (>16,000) of the annotated protein-coding genes in humans, we obtained novel information at multiple levels: novel protein-coding genes, unannotated exons, novel splice sites, proof of translation of pseudogenes (i.e. genes incorrectly annotated as pseudogenes), fused genes, SNPs encoded in proteins and novel N-termini to name a few. Many proteins identified in this study were identified by proteomic methods for the first time (e.g. hypothetical proteins or proteins annotated based solely on their chromosomal location). We have generated a catalog of proteins that show a more tissue-restricted pattern of expression, which should serve as the basis for pursuing biomarkers for diseases pertaining to specific organs. This study also provides one of the largest sets of proteotypic peptides for use in developing MRM assays for human proteins. Identification of several novel protein-coding regions in the human genome underscores the importance of systematic characterization of the human proteome and accurate annotation of protein-coding genes. This comprehensive dataset will complement other global HUPO initiatives using antibody-based as well as MRM mass spectrometry-based strategies. Finally, we believe that this dataset will become a reference set for use as a spectral library as well as for interesting interrogations pertaining to biomedical as well as bioinformatics questions.

Akhilesh (2)

Delegate Talk: Proteomic profiling of gallbladder cancer secretome – a source for circulatory biomarker discovery @ Amriteshwari Hall
Aug 14 @ 12:55 pm – 1:06 pm
Delegate Talk: Proteomic profiling of gallbladder cancer secretome – a source for circulatory biomarker discovery @ Amriteshwari Hall | Vallikavu | Kerala | India

Tejaswini Subbannayya, Nandini A. Sahasrabuddhe, Arivusudar Marimuthu, Santosh Renuse, Gajanan Sathe, Srinivas M. Srikanth, Mustafa A. Barbhuiya, Bipin Nair, Juan Carlos Roa, Rafael Guerrero-Preston, H. C. Harsha, David Sidransky, Akhilesh Pandey, T. S. Keshava Prasad and Aditi Chatterjee


Proteomic profiling of gallbladder cancer secretome – a source for circulatory biomarker discovery

Gallbladder cancer (GBC) is the fifth most common cancer of the gastrointestinal tract and one of the common malignancies that occur in the biliary tract (Misra et al. 2006; Lazcano-Ponce et al. 2001). It has a poor prognosis with survival of less than 5 years in 90% of the cases (Misra et al. 2003). The etiology is ill-defined. Several risk factors have been reported including cholelithiasis, obesity, female gender and exposure to carcinogens (Eslick 2010; Kumar et al. 2006). Poor prognosis in GBC is mainly due to late presentation of the disease and lack of reliable biomarkers for early diagnosis. This emphasizes the need to identify and characterize cancer biomarkers to aid in the diagnosis and prognosis of GBC. Secreted proteins are an important class of molecules which can be detected in body fluids and has been targeted for biomarker discovery. There are challenges faced in the proteomic interrogation of body fluids especially plasma such as low abundance of tumor secreted proteins, high complexity and high abundance of other proteins that are not released by the tumor cells (Tonack et al. 2009). Profiling of conditioned media from the cancer cell lines can be used as an alternate means to identify secreted proteins from tumor cells (Kashyap et al. 2010; Marimuthu et al. 2012). We analyzed the invasive property of 7 GBC cell lines (SNU-308, G-415, GB-d1, TGBC2TKB, TGBC24TKB, OCUG-1 and NOZ). Four cell lines were selected for analysis of the cancer secretome based on the invasive property of the cells. We employed isobaric tags for relative and absolute quantitation (iTRAQ) labeling technology coupled with high resolution mass spectrometry to identify and characterize secretome from the panel of 4GBC cancer cells mentioned above. In total, we have identified around 2,000 proteins of which 175 were secreted at differential abundance across all the four cell lines. This secretome analysis will act as a reservoir of candidate biomarkers. Currently, we are investigating and validating these candidate markers from GBC cell secretome. Through this study, we have shown mass spectrometry-based quantitative proteomic analysis as a robust approach to investigate secreted proteins in cancer cells.