Aug
12
Mon
2013
Invited Talk: Screening flavonoids for NF-kB inhibitory effect as potential breast cancer therapy @ Sathyam Hall
Aug 12 @ 11:00 am – 11:20 am

ayyappanAyyappan Nair, Ph.D.
Head, Business Development (Technologies, Discovery Biology), Anthem Biosciences & DavosPharma, New Jersey, USA


Inhibition of NF-κB regulated gene expression by chrysoeriol suppresses tumorigenesis in breast cancer cells

Amrutha K1, Pandurangan Nanjan1, Sanu K Shaji1, Damu Sunilkumar1, Subhalakshmi K1, Rashmi U Nair1, Lakshmi Rajakrishna2, Asoke Banerji1, Ayyappan Ramesh Nair1*,2

  1. School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri Campus, Clappana P.O., Kollam – 690 525, Kerala, India
  2. Anthem Biosciences, No 49, Canara Bank Road, Bommasandra Industrial Area, Phase 1,  Hosur Road, Bangalore – 560 099, Karnataka, India

Abstract:  A large number of effective cancer-preventing compounds inhibit the activation of nuclear factor-κ B (NF-κB).  It has been previously demonstrated that some flavonoids that are a vital component of our diet inhibits this pathway. As a consequence, many flavonoids inhibit genes involved in various aspects of tumorigenesis and have thus emerged as potential chemopreventive candidates for cancer treatment. We studied the effect of 17 different flavonoids, including the highly evaluated quercetin on the NF-κB pathway, and on the expression of MMP-9 and COX-2 (two NF-κB regulated genes involved in metastasis) in the highly invasive human breast cancer cell line MDA-MB-231.  The findings suggest that not all the quercetin like flavone backbone compounds inhibit the NF-κB pathway, and that the highly hydoxylated flavonols quercetagetin and gossypetin did not inhibit this pathway, nor did it inhibit the expression of MMP-9 and COX-2.  This indicates a correlation between inhibition of NF-κB and subsequent suppression of these NF-κB regulated genes. Here, we also report the novel observation that the not so well characterized methoxylated flavone chrysoeriol inhibited the NF-κB pathway, and was most potent in reducing the expression of MMP-9 and COX-2.  Based on these observations, the cellular effects of chrysoeriol were evaluated in MDA-MB-231.  Chrysoeriol caused cell cycle arrest at G2/M, inhibited migration and invasion, and caused cell death of macrophages that contributed to migration of these cancer cells.  These effects of chrysoeriol make it a potential therapeutic candidate for breast cancer metastasis.

Ayyappan

 

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
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: Regulation of the MHC complex and HLA solubilisation by the Flavivirus, Japanese Encephalitis Virus @ Acharya Hall
Aug 13 @ 12:13 pm – 12:40 pm

ManjunathR. Manjunath, Ph.D.
Associate Professor, Dept of Biochemistry, Indian Institute of Science, Bengaluru, India


REGULATION OF THE MHC COMPLEX AND HLA SOLUBILISATION BY THE FLAVIVIRUS, JAPANESE ENCEPHALITIS VIRUS

Viral encephalitis caused by Japanese encephalitis virus (JEV) and West Nile Virus (WNV) is a mosquito-borne disease that is prevalent in different parts of India and other parts of South East Asia. JEV is a positive single stranded RNA virus that belongs to the Flavivirus genus of the family Flaviviridae. The genome of JEV is about 11 kb long and codes for a polyprotein which is cleaved by both host and viral encoded proteases to form 3 structural and 7 non-structural proteins. It is a neurotropic virus which infects the central nervous system (CNS) and causes death predominantly in newborn children and young adults. JEV follows a zoonotic life-cycle involving mosquitoes and vertebrate, chiefly pigs and ardeid birds, as amplifying hosts. Humans are infected when bitten by an infected mosquito and are dead end hosts. Its structural, pathological, immunological and epidemiological aspects have been well studied. After entry into the host following a mosquito bite, JEV infection leads to acute peripheral neutrophil leucocytosis in the brain and leads to elevated levels of type I interferon, macrophage-derived chemotactic factor, RANTES,TNF-α and IL-8 in the serum and cerebrospinal fluid.

Major Histocompatibility Complex (MHC) molecules play a very important role in adaptive immune responses. Along with various classical MHC class I molecules, other non-classical MHC class I molecules play an important role in modulating innate immune responses. Our lab has shown the activation of cytotoxic T-cells (CTLs) during JEV infection and CTLs recognize non-self peptides presented on MHC molecules and provide protection by eliminating infected cells. However, along with proinflammatory cytokines such as TNFα, they may also cause immunopathology within the JEV infected brain. Both JEV and WNV, another related flavivirus have been shown to increase MHC class I expression. Infection of human foreskin fibroblast cells (HFF) by WNV results in upregulation of HLA expression. Data from our lab has also shown that JEV infection upregulates classical as well as nonclassical (class Ib) MHC antigen expression on the surface of primary mouse brain astrocytes and mouse embryonic fibroblasts.

There are no reports that have discussed the expression of these molecules on other cells like endothelial and astrocyte that play an important role in viral invasion in humans. We have studied the expression of human classical class I molecules HLA-A, -B, -C and the non-classical HLA molecules, HLA-E as well as HLA-F in immortalized human brain microvascular endothelial cells (HBMEC), human endothelial cell line (ECV304), human glioblastoma cell line (U87MG) and human foreskin fibroblast cells (HFF). Nonclassical MHC molecules such as mouse Qa-1b and its human homologue, HLA-E have been shown to be the ligand for the inhibitory NK receptor, NKG2A/CD94 and may bridge innate and adaptive immune responses. We show that JEV infection of HBMEC and ECV 304 cells upregulates the expression of HLA-A, and –B antigens as well as HLA-E and HLA-F. Increased expression of total HLA-E upon JEV infection was also observed in other human cell lines as well like, human amniotic epithelial cells, AV-3, FL and WISH cells. Further, we show for the first time that soluble HLA-E (sHLA-E) was released from infected ECV and HBMECs. In contrast, HFF cells showed only upregulation of cell-surface HLA-E expression while U87MG, a human glioblastoma cell line neither showed any cell-surface induction nor its solubilization. This shedding of sHLA-E was found to be dependent on matrix metalloproteinase (MMP) and an important MMP, MMP-9 was upregulated during JEV infection. Treatment with IFNγ resulted in the shedding of sHLA-E from ECV as well as U87MG but not from HFF cells. Also, sHLA-E was shed upon treatment with IFNβ and both IFNβ and TNFα, when present together caused an additive increase in the shedding of sHLA-E. HLA-E is an inhibitory ligand for CD94/NKG2A receptor of Natural Killer cells. Thus, MMP mediated solubilization of HLA-E from infected endothelial cells may have important implications in JEV pathogenesis including its ability to compromise the blood brain barrier.

Manjunath (2)

Invited Talk: The system of PAS proteins (HIF and AhR) as an interface between environment and skin homeostasis @ Acharya Hall
Aug 13 @ 2:33 pm – 2:50 pm

andreyAndrey Panteleyev, Ph.D.
Vice Chair, Division of Molecular Biology, NBICS Centre-Kurchatov Institute, Moscow, Russia


The system of PAS proteins (HIF and AhR) as an interface between environment and skin homeostasis

Regulation of normal skin functions as well as etiology of many skin diseases are both tightly linked to the environmental impact. Nevertheless, molecular aspects of skin-environment communication and mechanisms coordinating skin response to a plurality of environmental stressors remain poorly understood.

Our studies along with the work of other groups have identified the family of PAS dimeric transcription factors as an essential sensory and regulatory component of communication between skin and the environment. This protein family comprises a number of hypoxia-induced factors (HIF-alpha proteins), aryl hydrocarbon receptor (AhR), AhR nuclear translocator (ARNT), and several proteins implicated in control of rhythmic processes (Clock, Period, and Bmal proteins). Together, various PAS proteins (and first of all ARNT – as the central dimerization partner in the family) control such pivotal aspects of cell physiology as drug/xenobiotic metabolism, hypoxic and UV light response, ROS activity, pathogen defense, overall energy balance and breathing pathways.

In his presentation Dr. Panteleyev will focus on the role of ARNT activity and local hypoxia in control of keratinocyte differentiation and cornification. His recent work revealed that ARNT negatively regulates expression of late differentiation genes through modulation of amphiregulin expression and downstream alterations in activity of EGFR pathway. All these effects are highly dependent on epigenetic mechanisms such as histone deacetylation. Characterisation of hypoxia as a key microenvironmental factor in the skin and the role of HIF pathway in control of dermal vasculature and epidermal functions is another major focus of Dr. Panteleyev’s presentation.

In general, the studies of Dr. Panteleyev’s laboratory provide an insight into the PAS-dependent maintenance of skin homeostasis and point to the potential role of these proteins in pathogenesis of environmentally-modulated skin diseases such as barrier defects, desquamation abnormalities, psoriasis, etc.

 

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.