Aug
12
Mon
2013
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