406
Views
2
CrossRef citations to date
0
Altmetric
Review

The development and clinical applications of proteomics: an Indian perspective

ORCID Icon, , , , , & ORCID Icon show all
Pages 433-451 | Received 26 Mar 2020, Accepted 22 Jun 2020, Published online: 13 Jul 2020

References

  • Cobb M. 60 years ago, Francis Crick changed the logic of biology. PLoS Biol. 2017;15:e2003243.
  • Omenn GS, Lane L, Overall CM, et al. Progress on identifying and characterizing the human proteome: 2019 metrics from the HUPO human proteome project. J Proteome Res. 2019;18:4098–4107.
  • Piovesan A, Antonaros F, Vitale L, et al. Human protein-coding genes and gene feature statistics in 2019. BMC Res Notes. 2019;12:315.
  • Aebersold R, Agar JN, Amster IJ, et al. How many human proteoforms are there? Nat Chem Biol. 2018;14:206–214.
  • The Human Isoform Proteome. The human protein altas [ Internet]. [cited 2020 June 11]. Available from: https://www.proteinatlas.org/humanproteome/tissue/isoform
  • Ponomarenko EA, Poverennaya EV, Ilgisonis EV, et al. The size of the human proteome: the width and depth. Int J Anal Chem. 2016;2016:7436849.
  • Wilkins MR, Sanchez JC, Gooley AA, et al. Progress with proteome projects: why all proteins expressed by a genome should be identified and how to do it. Biotechnol Genet Eng Rev. 1996;13:19–50.
  • Durmaz AA, Karaca E, Demkow U, et al. Evolution of genetic techniques: past, present, and beyond. Guan X, editor. Biomed Res Int. 2015;2015:461524.
  • Magalhaes S, Goodfellow BJ, Nunes A. Aging and proteins: what does proteostasis have to do with age? Curr Mol Med. 2018;18:178–189.
  • Klaips CL, Jayaraj GG, Hartl FU. Pathways of cellular proteostasis in aging and disease. J Cell Biol. 2018;217:51–63.
  • Demichev V, Messner CB, Vernardis SI, et al. DIA-NN: neural networks and interference correction enable deep proteome coverage in high throughput. Nat Methods. 2020;17:41–44.
  • Kawashima Y, Watanabe E, Umeyama T, et al. Optimization of data-independent acquisition mass spectrometry for deep and highly sensitive proteomic analysis. Int J Mol Sci. 2019;20:5932.
  • Morsa D, Baiwir D, La Rocca R, et al. Multi-enzymatic limited digestion: the next-generation sequencing for proteomics? J Proteome Res. 2019;18:2501–2513.
  • Yu D, Wang Z, Cupp-Sutton KA, et al. Deep intact proteoform characterization in human cell lysate using high-pH and low-pH reversed-phase liquid chromatography. J Am Soc Mass Spectrom. 2019;30:2502–2513.
  • Yang L, George J, Wang J. Deep profiling of cellular heterogeneity by emerging single-cell proteomic technologies. Proteomics. 2019;1900226.
  • Jensen ON. Interpreting the protein language using proteomics. Nat Rev Mol Cell Biol. 2006;7:391–403.
  • McCormack AL, Schieltz DM, Goode B, et al. Direct analysis and identification of proteins in mixtures by LC/MS/MS and database searching at the low-femtomole level. Anal Chem. 1997;69:767–776.
  • Yates JR, Gilchrist A, Howell KE, et al. Proteomics of organelles and large cellular structures. Nat Rev Mol Cell Biol. 2005;6:702–714.
  • Yates JR III. Recent technical advances in proteomics. F1000Research. 2019;8:351.
  • Lippolis R, Angelis MD. Proteomics and human diseases. J Proteomics Bioinf. 2016;9:1–12.
  • Dorr KM, Conlon FL. Proteomic-based approaches to cardiac development and disease. Curr Opin Chem Biol. 2019;48:150–157.
  • Reddy PJ, Atak A, Ghantasala S, et al. Proteomics research in India: an update. J Proteomics. 2015;127:7–17.
  • Sirdeshmukh R. Indian proteomics efforts and human proteome project. J Proteomics. 2015;127:147–151.
  • Zingde SM. Has proteomics come of age in India? J Proteomics. 2015;127:3–6.
  • Priyadarshini S. India making a mark in global proteomics research. Nat India Special Issue. 2015:3–4.
  • Kim MS, Pinto SM, Getnet D, et al. A draft map of the human proteome. Nature. 2014;509:575–581.
  • Navani S. Manual evaluation of tissue microarrays in a high-throughput research project: the contribution of Indian surgical pathology to the Human Protein Atlas (HPA) project. Proteomics. 2016;16:1266–1270.
  • Chromosome-Centric Human Proteome Project (C-HPP) [ Internet]. [cited 2020 June 6]. Available from: https://www.hupo.org/C-HPP
  • Mukherjee S, Bandyopadhyay A. Proteomics in India: the clinical aspect. Clin Proteomics. 2016;13:21.
  • Crutchfield CA, Thomas SN, Sokoll LJ, et al. Advances in mass spectrometry-based clinical biomarker discovery. Clin Proteomics. 2016;13:1.
  • Banerjee S. Empowering clinical diagnostics with mass spectrometry. ACS Omega. 2020;5:2041–2048.
  • Ray S, Koshy NR, Reddy PJ, et al. Virtual Labs in proteomics: new e-learning tools. J Proteomics. 2012;75:2515–2525.
  • Ray S, Koshy NR, Diwakar S, et al. Sakshat labs: India’s virtual proteomics initiative. PLoS Biol. 2012;10:e1001353.
  • International Cancer Proteogenome Consortium | Office of Cancer Clinical Proteomics Research [ Internet]. [cited 2020 June 12]. Available from: https://proteomics.cancer.gov/programs/international-cancer-proteogenome-consortium
  • Bringans SD, Ito J, Stoll T, et al. Comprehensive mass spectrometry based biomarker discovery and validation platform as applied to diabetic kidney disease. EuPA Open Proteom. 2017;14:1–10.
  • Karkra K, Tetala KKR, Vijayalakshmi MA. A structure based plasma protein pre-fractionation using conjoint immobilized metal/chelate affinity (IMA) system. J Chromatogr B Analyt Technol Biomed Life Sci. 2017;1052:1–9.
  • Savane TS, Kumar S, Janakiraman VN, et al. Molecular insight in the purification of immunoglobulin by pseudobiospecific ligand l-histidine and histidyl moieties in histidine ligand affinity chromatography (HLAC) by molecular docking. J Chromatogr B Analyt Technol Biomed Life Sci. 2016;1021:129–136.
  • Prasanna RR, Kamalanathan AS, Vijayalakshmi MA. Development of L-histidine immobilized CIM(®) monolithic disks for purification of immunoglobulin G. J Mol Recognit. 2015;28:129–141.
  • Prasanna RR, Sidhik S, Kamalanathan AS, et al. Affinity selection of histidine-containing peptides using metal chelate methacrylate monolithic disk for targeted LC-MS/MS approach in high-throughput proteomics. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;955–956:42–49.
  • Yan JX, Devenish AT, Wait R, et al. Fluorescence two-dimensional difference gel electrophoresis and mass spectrometry based proteomic analysis of Escherichia coli. Proteomics. 2002;2:1682–1698.
  • Dutta M, Subramani E, Taunk K, et al. Investigation of serum proteome alterations in human endometriosis. J Proteomics. 2015;114:182–196.
  • Gollapalli K, Ray S, Srivastava R, et al. Investigation of serum proteome alterations in human glioblastoma multiforme. Proteomics. 2012;12:2378–2390.
  • Gopalakrishnan V, Purushothaman P, Bhaskar A. Proteomic analysis of plasma proteins in diabetic retinopathy patients by two dimensional electrophoresis and MALDI-Tof-MS. J Diabetes Complications. 2015;29:928–936.
  • Mallikarjuna K, Sundaram CS, Sharma Y, et al. Comparative proteomic analysis of differentially expressed proteins in primary retinoblastoma tumors. Proteomics Clin Appl. 2010;4:449–463.
  • Rukmangadachar LA, Kataria J, Hariprasad G, et al. Two-dimensional difference gel electrophoresis (DIGE) analysis of sera from visceral leishmaniasis patients. Clin Proteomics. 2011;8:4.
  • Saijyothi AV, Angayarkanni N, Syama C, et al. Two dimensional electrophoretic analysis of human tears: collection method in dry eye syndrome. Electrophoresis. 2010;31:3420–3427.
  • Ong SE, Blagoev B, Kratchmarova I, et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics. 2002;1:376–386.
  • Dayon L, Hainard A, Licker V, et al. Relative quantification of proteins in human cerebrospinal fluids by MS/MS using 6-plex isobaric tags. Anal Chem. 2008;80:2921–2931.
  • Ross PL, Huang YN, Marchese JN, et al. Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol Cell Proteomics. 2004;3:1154–1169.
  • Chanukuppa V, Paul D, Taunk K, et al. XPO1 is a critical player for bortezomib resistance in multiple myeloma: A quantitative proteomic approach. J Proteomics. 2019;209:103504.
  • Gautam P, Nair SC, Gupta MK, et al. Proteins with altered levels in plasma from glioblastoma patients as revealed by iTRAQ-based quantitative proteomic analysis. PloS One. 2012;7:e46153.
  • Gollapalli K, Ghantasala S, Atak A, et al. Tissue proteome analysis of different grades of human gliomas provides major cues for glioma pathogenesis. OMICS. 2017;21:275–284.
  • Paul D, Chanukuppa V, Reddy PJ, et al. Global proteomic profiling identifies etoposide chemoresistance markers in non-small cell lung carcinoma. J Proteomics. 2016;138:95–105.
  • Sharma S, Ray S, Moiyadi A, et al. Quantitative proteomic analysis of meningiomas for the identification of surrogate protein markers. Sci Rep. 2014;4:7140.
  • Lai T, Yang Y, Ng SK. Advances in mammalian cell line development technologies for recombinant protein production. Pharmaceuticals (Basel). 2013;6:579–603.
  • Ali SA, Kaur G, Kaushik JK, et al. Examination of pathways involved in leukemia inhibitory factor (LIF)-induced cell growth arrest using label-free proteomics approach. J Proteomics. 2017;168:37–52.
  • Mary S, Kulkarni MJ, Malakar D, et al. Placental proteomics provides insights into pathophysiology of pre-eclampsia and predicts possible markers in plasma. J Proteome Res. 2017;16:1050–1060.
  • Mukherjee S, Jagadeeshaprasad MG, Banerjee T, et al. Proteomic analysis of human plasma in chronic rheumatic mitral stenosis reveals proteins involved in the complement and coagulation cascade. Clin Proteomics. 2014;11:35.
  • Shi T, Song E, Nie S, et al. Advances in targeted proteomics and applications to biomedical research. Proteomics. 2016;16:2160–2182.
  • Dillen L, Cools W, Vereyken L, et al. Comparison of triple quadrupole and high-resolution TOF-MS for quantification of peptides. Bioanalysis. 2012;4:565–579.
  • Shi T, Su D, Liu T, et al. Advancing the sensitivity of selected reaction monitoring-based targeted quantitative proteomics. Proteomics. 2012;12:1074–1092.
  • Peterson AC, Russell JD, Bailey DJ, et al. Parallel reaction monitoring for high resolution and high mass accuracy quantitative, targeted proteomics. Mol Cell Proteomics. 2012;11:1475–1488.
  • Gillet LC, Navarro P, Tate S, et al. Targeted data extraction of the MS/MS spectra generated by data-independent acquisition: a new concept for consistent and accurate proteome analysis. Mol Cell Proteomics. 2012;11:O111 016717.
  • Liu Y, Huttenhain R, Collins B, et al. Mass spectrometric protein maps for biomarker discovery and clinical research. Expert Rev Mol Diagn. 2013;13:811–825.
  • McDonnell LA, Heeren RM. Imaging mass spectrometry. Mass Spectrom Rev. 2007;26:606–643.
  • Walch A, Rauser S, Deininger SO, et al. MALDI imaging mass spectrometry for direct tissue analysis: a new frontier for molecular histology. Histochem Cell Biol. 2008;130:421–434.
  • Delehanty JB, Ligler FS. Method for printing functional protein microarrays. BioTechniques. 2003;34:380–385.
  • Zhu H, Bilgin M, Bangham R, et al. Global analysis of protein activities using proteome chips. Science. 2001;293:2101–2105.
  • Sutandy FX, Qian J, Chen CS, et al. Overview of protein microarrays. Curr Protoc Protein Sci. 2013;72:27.1.1–27.1.16. Chapter 27:Unit 27 1.
  • Huang Y, Zhu H. Protein array-based approaches for biomarker discovery in cancer. Genomics Proteomics Bioinf. 2017;15:73–81.
  • Pawar H, Kashyap MK, Sahasrabuddhe NA, et al. Quantitative tissue proteomics of esophageal squamous cell carcinoma for novel biomarker discovery. Cancer Biol Ther. 2011;12:510–522.
  • Sahasrabuddhe NA, Barbhuiya MA, Bhunia S, et al. Identification of prosaposin and transgelin as potential biomarkers for gallbladder cancer using quantitative proteomics. Biochem Biophys Res Commun. 2014;446:863–869.
  • Kumar D, Bansal G, Narang A, et al. Integrating transcriptome and proteome profiling: strategies and applications. Proteomics. 2016;16:2533–2544.
  • Kumar D, Yadav AK, Dash D. Choosing an optimal database for protein identification from tandem mass spectrometry data. Methods Mol Biol. 2017;1549:17–29. New York, NY: Springer.
  • Kumar D, Dash D. Proteogenomic tools and approaches to explore protein coding landscapes of eukaryotic genomes. Adv Exp Med Biol. 2016;926:1–10.
  • Kumar D, Yadav AK, Jia X, et al. Integrated transcriptomic-proteomic analysis using a proteogenomic workflow refines rat genome annotation. Mol Cell Proteomics. 2016;15:329–339.
  • Mathivanan S, Ahmed M, Ahn NG, et al. Human proteinpedia enables sharing of human protein data. Nat Biotechnol. 2008;26:164–167.
  • Kandasamy K, Mohan SS, Raju R, et al. NetPath: a public resource of curated signal transduction pathways. Genome Biol. 2010;11:R3.
  • Agarwal SM, Raghav D, Singh H, et al. CCDB: a curated database of genes involved in cervix cancer. Nucleic Acids Res. 2011;39:D975–9.
  • Tyagi A, Tuknait A, Anand P, et al. CancerPPD: a database of anticancer peptides and proteins. Nucleic Acids Res. 2015;43:D837–43.
  • Gautam A, Chaudhary K, Singh S, et al. Hemolytik: a database of experimentally determined hemolytic and non-hemolytic peptides. Nucleic Acids Res. 2014;42:D444–9.
  • Choubey J, Choudhari JK, Patel A, et al. Respiratory cancer database: an open access database of respiratory cancer gene and miRNA. J Cancer Res Ther. 2017;13:487–490.
  • Sivakumaran S, Hariharaputran S, Mishra J, et al. The database of quantitative cellular signaling: management and analysis of chemical kinetic models of signaling networks. Bioinformatics. 2003;19:408–415.
  • Balaji S, Sujatha S, Kumar SSC, et al. PALI—a database of phylogeny and alignment of homologous protein structures. Nucleic Acids Res. 2001;29:61–65.
  • Das AA, Sharma OP, Kumar MS, et al. PepBind: a comprehensive database and computational tool for analysis of protein–peptide interactions. Genomics Proteomics Bioinf. 2013;11:241–246.
  • Gowri VS, Krishnadev O, Swamy CS, et al. MulPSSM: a database of multiple position-specific scoring matrices of protein domain families. Nucleic Acids Res. 2006;34:D243–6.
  • Jadhav A, Ezhilarasan V, Sharma OP, et al. Clostridium-DTDB: a comprehensive database for potential drug targets of Clostridium difficile. Comput Biol Med. 2013;43:362–367.
  • Krishnadev O, Rekha N, Pandit SB, et al. PRODOC: a resource for the comparison of tethered protein domain architectures with in-built information on remotely related domain families. Nucleic Acids Res. 2005;33:W126–W129.
  • Krupa A, Abhinandan KR, Srinivasan N. KinG: a database of protein kinases in genomes. Nucleic Acids Res. 2004;32:D153–5.
  • Mudgal R, Sandhya S, Kumar G, et al. NrichD database: sequence databases enriched with computationally designed protein-like sequences aid in remote homology detection. Nucleic Acids Res. 2015;43:D300–5.
  • Sharma O, Das A, Krishna R, et al. Structural epitope database (SEDB): a web-based database for the epitope, and its intermolecular interaction along with the tertiary structure information. J Proteomics Bioinf. 2012;5:084–089.
  • Sharma OP, Jadhav A, Hussain A, et al. VPDB: viral protein structural database. Bioinformation. 2011;6:324.
  • Gadewal NS, Zingde SM. Database and interaction network of genes involved in oral cancer: version II. Bioinformation. 2011;6:169–170.
  • Gadewal NS, Zingde SM. Database and interactome map of genes involved in oral cancer. OJB. 2007;8:41–44.
  • Bhartiya D, Kumar A, Singh H, et al. OrCanome: a comprehensive resource for oral cancer. Asian Pac J Cancer Prev. 2016;17:1333–1336.
  • The India State-Level Disease Burden Initiative | Public Health Foundation of India [ Internet]. 2017 [cited 2020 June 6]. Available from: https://phfi.org/the-work/research/the-india-state-level-disease-burden-initiative/
  • Lifestyle diseases in India [ Internet]. 2018 [cited 2020 June 6]. Available from: https://pib.gov.in/Pressreleaseshare.aspx?PRID=1540840
  • Gahoi N, Malhotra D, Moiyadi A, et al. Multi-pronged proteomic analysis to study the glioma pathobiology using cerebrospinal fluid samples. Proteomics – Clin Appl. 2018;12:1700056.
  • Gollapalli K, Ghantasala S, Kumar S, et al. Subventricular zone involvement in glioblastoma - a proteomic evaluation and clinicoradiological correlation. Sci Rep. 2017;7:1449.
  • Jain R, Atak A, Yeola A, et al. Proteomic level changes associated with S3I201 treated U87 glioma cells. J Proteomics. 2017;150:341–350.
  • Syed P, Gupta S, Choudhary S, et al. Autoantibody profiling of glioma serum samples to identify biomarkers using human proteome arrays. Sci Rep. 2015;5:13895.
  • Jain R, Kulkarni P, Dhali S, et al. Quantitative proteomic analysis of global effect of LLL12 on U87 cell’s proteome: an insight into the molecular mechanism of LLL12. J Proteomics. 2015;113:127–142.
  • Chumbalkar VC, Subhashini C, Dhople VM, et al. Differential protein expression in human gliomas and molecular insights. Proteomics. 2005;5:1167–1177.
  • Polisetty RV, Gautam P, Sharma R, et al. LC-MS/MS analysis of differentially expressed glioblastoma membrane proteome reveals altered calcium signaling and other protein groups of regulatory functions. Mol Cell Proteomics. 2012;11:M111.013565.
  • Polisetty RV, Gautam P, Gupta MK, et al. Heterogeneous nuclear ribonucleoproteins and their interactors are a major class of deregulated proteins in anaplastic astrocytoma: a grade III malignant glioma. J Proteome Res. 2013;12:3128–3138.
  • Jayaram S, Gupta MK, Polisetty RV, et al. Towards developing biomarkers for glioblastoma multiforme: a proteomics view. Expert Rev Proteomics. 2014;11:621–639.
  • Gupta MK, Polisetty RV, Sharma R, et al. Altered transcriptional regulatory proteins in glioblastoma and YBX1 as a potential regulator of tumor invasion. Sci Rep. 2019;9:10986.
  • Nijaguna MB, Patil V, Urbach S, et al. Glioblastoma-derived macrophage colony-stimulating factor (MCSF) induces microglial release of insulin-like growth factor-binding protein 1 (IGFBP1) to promote angiogenesis. J Biol Chem. 2015;290:23401–23415.
  • Kumar DM, Patil V, Ramachandran B, et al. Temozolomide-modulated glioma proteome: role of interleukin-1 receptor-associated kinase-4 (IRAK4) in chemosensitivity. Proteomics. 2013;13:2113–2124.
  • Kumar DM, Thota B, Shinde SV, et al. Proteomic identification of haptoglobin α2 as a glioblastoma serum biomarker: implications in cancer cell migration and tumor growth. J Proteome Res. 2010;9:5557–5567.
  • Somasundaram K, Nijaguna MB, Kumar DM. Serum proteomics of glioma: methods and applications. Expert Rev Mol Diagn. 2009;9:695–707.
  • Arora A, Patil V, Kundu P, et al. Serum biomarkers identification by iTRAQ and verification by MRM: S100A8/S100A9 levels predict tumor-stroma involvement and prognosis in glioblastoma. Sci Rep. 2019;9:2749.
  • Gajbhiye A, Dabhi R, Taunk K, et al. Multipronged quantitative proteomics reveals serum proteome alterations in breast cancer intrinsic subtypes. J Proteomics. 2017;163:1–13.
  • Pendharkar N, Gajbhiye A, Taunk K, et al. Quantitative tissue proteomic investigation of invasive ductal carcinoma of breast with luminal B HER2 positive and HER2 enriched subtypes towards potential diagnostic and therapeutic biomarkers. J Proteomics. 2016;132:112–130.
  • Gajbhiye A, Dabhi R, Taunk K, et al. Urinary proteome alterations in HER2 enriched breast cancer revealed by multipronged quantitative proteomics. Proteomics. 2016;16:2403–2418.
  • Korwar AM, Bhonsle HS, Ghole VS, et al. Proteomic profiling and interactome analysis of ER-positive/HER2/neu negative invasive ductal carcinoma of the breast: towards proteomics biomarkers. OMICS. 2013;17:27–40.
  • Giri K, Mehta A, Ambatipudi K. In search of the altering salivary proteome in metastatic breast and ovarian cancers. FASEB Bioadv. 2019;1:191–207.
  • Solanki HS, Raja R, Zhavoronkov A, et al. Targeting focal adhesion kinase overcomes erlotinib resistance in smoke induced lung cancer by altering phosphorylation of epidermal growth factor receptor. Oncoscience. 2018;5:21–38.
  • Babu N, Advani J, Solanki HS, et al. miRNA and proteomic dysregulation in non-small cell lung cancer in response to cigarette smoke. MicroRNA. 2018;7:38–53.
  • Solanki HS, Advani J, Khan AA, et al. Chronic cigarette smoke mediated global changes in lung mucoepidermoid cells: a phosphoproteomic analysis. OMICS. 2017;21:474–487.
  • Raja R, Sahasrabuddhe NA, Radhakrishnan A, et al. Chronic exposure to cigarette smoke leads to activation of p21 (RAC1)-activated kinase 6 (PAK6) in non-small cell lung cancer cells. Oncotarget. 2016;7:61229–61245.
  • Sathe G, Pinto SM, Syed N, et al. Phosphotyrosine profiling of curcumin-induced signaling. Clin Proteomics. 2016;13:13.
  • Govekar RB, D’Cruz AK, Alok Pathak K, et al. Proteomic profiling of cancer of the gingivo-buccal complex: identification of new differentially expressed markers. Proteomics Clin Appl. 2009;3:1451–1462.
  • Fulzele A, Malgundkar SA, Govekar RB, et al. Proteomic profile of keratins in cancer of the gingivo buccal complex: consolidating insights for clinical applications. J Proteomics. 2013;91:242–258.
  • Shukla S, Govekar RB, Sirdeshmukh R, et al. Tumor antigens eliciting autoantibody response in cancer of gingivo-buccal complex. Proteomics Clin Appl. 2007;1:1592–1604.
  • Shukla S, Pranay A, D’Cruz AK, et al. Immunoproteomics reveals that cancer of the tongue and the gingivobuccal complex exhibit differential autoantibody response. Cancer Biomark. 2009;5:127–135.
  • Pranay A, Shukla S, Kannan S, et al. Prognostic utility of autoantibodies to α-enolase and Hsp70 for cancer of the gingivo-buccal complex using immunoproteomics. Proteomics Clin Appl. 2013;7:392–402.
  • Sivadasan P, Gupta MK, Sathe G, et al. Salivary proteins from dysplastic leukoplakia and oral squamous cell carcinoma and their potential for early detection. J Proteomics. 2020;212:103574.
  • Ananthi S, Lakshmi CNP, Atmika P, et al. Global quantitative proteomics reveal deregulation of cytoskeletal and apoptotic signalling proteins in oral tongue squamous cell carcinoma. Sci Rep. 2018;8:1567.
  • Dey KK, Pal I, Bharti R, et al. Identification of RAB2A and PRDX1 as the potential biomarkers for oral squamous cell carcinoma using mass spectrometry-based comparative proteomic approach. Tumour Biol. 2015;36:9829–9837.
  • Chanukuppa V, Paul D, Taunk K, et al. Proteomics and functional study reveal marginal zone B and B1 cell specific protein as a candidate marker of multiple myeloma. Int J Oncol. 2020;57:325–337.
  • Kalra RS, Bapat SA. Expression proteomics predicts loss of RXR-γ during progression of epithelial ovarian cancer. Plos One. 2013;8:e70398.
  • Kalra RS, Bapat SA. Proteomics to predict loss of RXR-γ during progression of epithelial ovarian cancer. In: Ray SK, editor. Retinoid and rexinoid signaling: methods and protocols [ Internet]. New York (NY): Springer; 2019 [cited 2020 Mar 26]. p. 1–14.
  • Kaul D, Kaur R, Baba I, et al. Functional proteomics of receptor-Ck in the developmental stages of human atherosclerotic arterial wall. Indian Heart J. 2003;55:252–255.
  • Basak T, Tanwar VS, Bhardwaj G, et al. Plasma proteomic analysis of stable coronary artery disease indicates impairment of reverse cholesterol pathway. Sci Rep. 2016;6:28042.
  • Ghatge M, Sharma A, Maity S, et al. Danger-recognizing proteins, β-defensin-128 and histatin-3, as potential biomarkers of recurrent coronary events. Int J Mol Med. 2017;40:531–538.
  • Chowdhury D, Tangutur AD, Khatua TN, et al. A proteomic view of isoproterenol induced cardiac hypertrophy: prohibitin identified as a potential biomarker in rats. J Transl Med. 2013;11:130.
  • Datta K, Basak T, Varshney S, et al. Quantitative proteomic changes during post myocardial infarction remodeling reveals altered cardiac metabolism and desmin aggregation in the infarct region. J Proteomics. 2017;152:283–299.
  • Mitra A, Basak T, Ahmad S, et al. Comparative proteome profiling during cardiac hypertrophy and myocardial infarction reveals altered glucose oxidation by differential activation of pyruvate dehydrogenase E1 component subunit β. J Mol Biol. 2015;427:2104–2120.
  • Banarjee R, Sharma A, Bai S, et al. Proteomic study of endothelial dysfunction induced by AGEs and its possible role in diabetic cardiovascular complications. J Proteomics. 2018;187:69–79.
  • Kesavan SK, Bhat S, Golegaonkar SB, et al. Proteome wide reduction in AGE modification in streptozotocin induced diabetic mice by hydralazine mediated transglycation. Sci Rep. 2013;3:1–10.
  • Korwar AM, Vannuruswamy G, Jagadeeshaprasad MG, et al. Development of diagnostic fragment ion library for glycated peptides of human serum albumin: targeted quantification in prediabetic, diabetic, and microalbuminuria plasma by parallel reaction monitoring, SWATH, and MSE. Mol Cell Proteomics. 2015;14:2150–2159.
  • Jagadeeshaprasad MG, Batkulwar KB, Meshram NN, et al. Targeted quantification of N-1-(carboxymethyl) valine and N-1-(carboxyethyl) valine peptides of β-hemoglobin for better diagnostics in diabetes. Clin Proteomics. 2016;13:7.
  • Rathore R, Sonwane BP, Jagadeeshaprasad MG, et al. Glycation of glucose sensitive lysine residues K36, K438 and K549 of albumin is associated with prediabetes. J Proteomics. 2019;208:103481.
  • Jagadeeshaprasad MG, Venkatasubramani V, Unnikrishnan AG, et al. Albumin abundance and its glycation status determine hemoglobin glycation. ACS Omega. 2018;3:12999–13008.
  • Karthik D, Vijayakumar R, Pazhanichamy K, et al. A proteomics approach to identify the differential protein level in cardiac muscle of diabetic rat. Acta Biochim Pol. 2014;61:285–293.
  • Marikanty RK, Gupta MK, Cherukuvada SVB, et al. Identification of urinary proteins potentially associated with diabetic kidney disease. Indian J Nephrol. 2016;26:434–445.
  • Bhattacharjee M, Balakrishnan L, Renuse S, et al. Synovial fluid proteome in rheumatoid arthritis. Clin Proteomics. 2016;13:12.
  • Balakrishnan L, Bhattacharjee M, Ahmad S, et al. Differential proteomic analysis of synovial fluid from rheumatoid arthritis and osteoarthritis patients. Clin Proteomics. 2014;11:1.
  • Bhattacharjee M, Sharma R, Goel R, et al. A multilectin affinity approach for comparative glycoprotein profiling of rheumatoid arthritis and spondyloarthropathy. Clin Proteomics. 2013;10:11.
  • Saroha A, Biswas S, Chatterjee BP, et al. Altered glycosylation and expression of plasma alpha-1-acid glycoprotein and haptoglobin in rheumatoid arthritis. J Chromatogr B Analyt Technol Biomed Life Sci. 2011;879:1839–1843.
  • Saroha A, Kumar S, Chatterjee BP, et al. Jacalin bound plasma O-glycoproteome and reduced sialylation of alpha 2-HS glycoprotein (A2HSG) in rheumatoid arthritis patients. PLoS ONE. 2012;7:e46374.
  • Rai P, Kota V, Deendayal M, et al. Differential proteome profiling of eutopic endometrium from women with endometriosis to understand etiology of endometriosis. J Proteome Res. 2010;9:4407–4419.
  • Ghosh D, Nagpal S, Bhat MA, et al. Gel-free proteomics reveals neoplastic potential in endometrium of infertile patients with stage IV ovarian endometriosis. J Reprod Health Med. 2015;2:83–95.
  • Rashid N, Nigam A, Jain SK, et al. Proteomic sift through serum and endometrium profiles unraveled signature proteins associated with subdued fertility and dampened endometrial receptivity in women with polycystic ovary syndrome. Cell Tissue Res. 2020. DOI:10.1007/s00441-020-03171-3
  • Patil K, Yelamanchi S, Kumar M, et al. Quantitative mass spectrometric analysis to unravel glycoproteomic signature of follicular fluid in women with polycystic ovary syndrome. PLoS ONE. 2019;14:e0214742.
  • Ambekar AS, Kelkar DS, Pinto SM, et al. Proteomics of follicular fluid from women with polycystic ovary syndrome suggests molecular defects in follicular development. J Clin Endocrinol Metab. 2015;100:744–753.
  • Mary S, Kulkarni MJ, Mehendale SS, et al. Differential accumulation of vimentin fragments in preeclamptic placenta. Cytoskeleton (Hoboken). 2017;74:420–425.
  • Mary S, Kulkarni MJ, Mehendale SS, et al. Tubulointerstitial nephritis antigen-like 1 protein is downregulated in the placenta of pre-eclamptic women. Clin Proteomics. 2017;14:8.
  • Sahar T, Nigam A, Anjum S, et al. Interactome Analysis of the differentially expressed proteins in uterine leiomyoma. Anticancer Agents Med Chem. 2019;19:1293–1312.
  • Rawat P, Bathla S, Baithalu R, et al. Identification of potential protein biomarkers for early detection of pregnancy in cow urine using 2D DIGE and label free quantitation. Clin Proteomics. 2016;13:15.
  • Mohanty G, Jena SR, Nayak J, et al. Quantitative proteomics decodes clusterin as a critical regulator of paternal factors responsible for impaired compensatory metabolic reprogramming in recurrent pregnancy loss. Andrologia. 2020;52:e13498.
  • Tomar AK, Sooch BS, Raj I, et al. Isolation and identification of concanavalin A binding glycoproteins from human seminal plasma: a step towards identification of male infertility marker proteins. Dis Markers. 2011;31:379–386.
  • Tomar AK, Sooch BS, Singh S, et al. Differential proteomics of human seminal plasma: A potential target for searching male infertility marker proteins. Proteomics Clin Appl. 2012;6:147–151.
  • Yadav VK, Kumar V, Chhikara N, et al. Purification and characterization of a native zinc-binding high molecular weight multiprotein complex from human seminal plasma. J Sep Sci. 2011;34:1076–1083.
  • Kant K, Tomar AK, Singh S, et al. Ageing associated proteomic variations in seminal plasma of Indian men. J Proteins Proteomics. 2019;10:83–89.
  • Mohanty G, Jena SR, Nayak J, et al. Proteomic signatures in spermatozoa reveal the role of paternal factors in recurrent pregnancy loss. World J Mens Health. 2020;38:103–114.
  • Sinha A, Singh V, Singh S, et al. Proteomic analyses reveal lower expression of TEX40 and ATP6V0A2 proteins related to calcium ion entry and acrosomal acidification in asthenozoospermic males. Life Sci. 2019;218:81–88.
  • Singh M, Mukherjee P, Narayanasamy K, et al. Proteome analysis of plasmodium falciparum extracellular secretory antigens at asexual blood stages reveals a cohort of proteins with possible roles in immune modulation and signaling. Mol Cell Proteomics. 2009;8:2102–2118.
  • Acharya P, Pallavi R, Chandran S, et al. Clinical proteomics of the neglected human malarial parasite plasmodium vivax. Plos One. 2011;6:e26623.
  • Venkatesh A, Lahiri A, Reddy PJ, et al. Identification of highly expressed plasmodium vivax proteins from clinical isolates using proteomics. Proteomics Clin Appl. 2018;12:e1700046.
  • Venkatesh A, Aggarwal S, Kumar S, et al. Comprehensive proteomics investigation of P. vivax-infected human plasma and parasite isolates. BMC Infect Dis. 2020;20:188.
  • Ranjan R, Chugh M, Kumar S, et al. Proteome analysis reveals a large merozoite surface protein-1 associated complex on the Plasmodium falciparum merozoite surface. J Proteome Res. 2011;10:680–691.
  • Ray S, Renu D, Srivastava R, et al. Proteomic investigation of falciparum and vivax malaria for identification of surrogate protein markers. Plos One. 2012;7:e41751.
  • Ray S, Patel SK, Venkatesh A, et al. Clinicopathological analysis and multipronged quantitative proteomics reveal oxidative stress and cytoskeletal proteins as possible markers for severe vivax malaria. Sci Rep. 2016;6:1–15.
  • Awasthi G, Tyagi S, Kumar V, et al. A proteogenomic analysis of haptoglobin in malaria. Proteomics Clin Appl. 2018;12:e1700077.
  • Ray S, Patel SK, Venkatesh A, et al. Quantitative proteomics analysis of plasmodium vivax induced alterations in human serum during the acute and convalescent phases of infection. Sci Rep. 2017;7:4400.
  • Jadhav M, Nayak M, Kumar S, et al. Clinical proteomics and cytokine profiling for dengue fever disease severity biomarkers. OMICS. 2017;21:665–677.
  • Ray S, Srivastava R, Tripathi K, et al. Serum proteome changes in dengue virus-infected patients from a dengue-endemic area of India: towards new molecular targets? OMICS. 2012;16:527–536.
  • Anbarasu D, Raja CP, Raja A. Multiplex analysis of cytokines/chemokines as biomarkers that differentiate healthy contacts from tuberculosis patients in high endemic settings. Cytokine. 2013;61:747–754.
  • Kumar B, Sharma D, Sharma P, et al. Proteomic analysis of Mycobacterium tuberculosis isolates resistant to kanamycin and amikacin. J Proteomics. 2013;94:68–77.
  • Tyagi P, Pal VK, Agrawal R, et al. Mycobacterium tuberculosis reactivates HIV-1 via exosome-mediated resetting of cellular redox potential and bioenergetics. mBio. 2020;11(2):e03293–19.
  • Arya R, Dabral D, Faruquee HM, et al. Serum small extracellular vesicles proteome of tuberculosis patients demonstrated deregulated immune response. Proteomics – Clin Appl. 2020;14:1900062.
  • Bishwal SC, Das MK, Badireddy VK, et al. Sputum proteomics reveals a shift in vitamin D-binding protein and antimicrobial protein axis in tuberculosis patients. Sci Rep. 2019;9:1036.
  • Kundu J, Verma A, Verma I, et al. Proteomic changes in Mycobacterium tuberculosis H37Rv under hyperglycemic conditions favour its growth through altered expression of Tgs3(Rv3234c) and supportive proteins (Rv0547c, AcrA1 and Mpa). Tuberculosis. 2019;115:154–160.
  • Ganji R, Dhali S, Rizvi A, et al. Understanding HIV-Mycobacteria synergism through comparative proteomics of intra-phagosomal mycobacteria during mono- and HIV co-infection. Sci Rep. 2016;6:22060.
  • Ganji R, Dhali S, Rizvi A, et al. Proteomics approach to understand reduced clearance of mycobacteria and high viral titers during HIV-mycobacteria co-infection. Cell Microbiol. 2016;18:355–368.
  • Dalal K, Dalal B, Bhatia S, et al. Analysis of serum haptoglobin using glycoproteomics and lectin immunoassay in liver diseases in hepatitis B virus infection. Clin Chim Acta. 2019;495:309–317.
  • Dalal K, Khorate P, Dalal B, et al. Differentially expressed serum host proteins in hepatitis B and C viral infections. Virusdisease. 2018;29:468–477.
  • Gouthamchandra K, Kumar A, Shwetha S, et al. Serum proteomics of hepatitis C virus infection reveals retinol-binding protein 4 as a novel regulator. J Gen Virol. 2014;95:1654–1667.
  • Taneja S, Sen S, Gupta VK, et al. Plasma and urine biomarkers in acute viral hepatitis E. Proteome Sci. 2009;7:39.
  • Taneja S, Ahmad I, Sen S, et al. Plasma peptidome profiling of acute hepatitis E patients by MALDI-TOF/TOF. Proteome Sci. 2011;9:5.
  • Kumar A, Singh S, Ahirwar SK, et al. Proteomics-based identification of plasma proteins and their association with the host-pathogen interaction in chronic typhoid carriers. Int J Infect Dis. 2014;19:59–66.
  • Garg G, Ali V, Singh K, et al. Quantitative secretome analysis unravels new secreted proteins in amphotericin B resistant Leishmania donovani. J Proteomics. 2019;207:103464.
  • Ejazi SA, Bhattacharyya A, Choudhury ST, et al. Immunoproteomic identification and characterization of Leishmania membrane proteins as non-invasive diagnostic candidates for clinical visceral leishmaniasis. Sci Rep. 2018;8:12110.
  • Singh N, Goel R, Jain E. Differential metabolic pathway analysis of the proteomes of Leishmania donovani and Leptomonas seymouri. Proteomics – Clin Appl. 2018;12:1600087.
  • Jamdhade MD, Pawar H, Chavan S, et al. Comprehensive proteomics analysis of glycosomes from Leishmania donovani. OMICS. 2015;19:157–170.
  • Singh AK, Pandey RK, Siqueira-Neto JL, et al. Proteomic-based approach to gain insight into reprogramming of THP-1 cells exposed to Leishmania donovani over an early temporal window. Infect Immun. 2015;83:1853–1868.
  • Kumar A, Misra P, Sisodia B, et al. Proteomic analyses of membrane enriched proteins of Leishmania donovani Indian clinical isolate by mass spectrometry. Parasitol Int. 2015;64:36–42.
  • Das R, Mitra G, Mathew B, et al. Mass spectrometry-based diagnosis of hemoglobinopathies: a potential tool for the screening of genetic disorder. Biochem Genet. 2016;54:816–825.
  • Karmakar S, Banerjee D, Chakrabarti A. Platelet proteomics in thalassemia: factors responsible for hypercoagulation. Proteomics Clin Appl. 2016;10:239–247.
  • Bhattacharya D, Saha S, Basu S, et al. Differential regulation of redox proteins and chaperones in HbEβ-thalassemia erythrocyte proteome. Proteomics Clin Appl. 2010;4:480–488.
  • Karmakar S, Saha S, Banerjee D, et al. Differential proteomics study of platelets in asymptomatic constitutional macrothrombocytopenia: altered levels of cytoskeletal proteins. Eur J Haematol. 2015;94:43–50.
  • Basu A, Saha S, Karmakar S, et al. 2D DIGE based proteomics study of erythrocyte cytosol in sickle cell disease: altered proteostasis and oxidative stress. Proteomics. 2013;13:3233–3242.
  • Saha S, Ramanathan R, Basu A, et al. Elevated levels of redox regulators, membrane-bound globin chains, and cytoskeletal protein fragments in hereditary spherocytosis erythrocyte proteome. Eur J Haematol. 2011;87:259–266.
  • Murthy KR, Goel R, Subbannayya Y, et al. Proteomic analysis of human vitreous humor. Clin Proteomics. 2014;11:29.
  • Danda R, Ganapathy K, Sathe G, et al. Membrane proteome of invasive retinoblastoma: differential proteins and biomarkers. Proteomics Clin Appl. 2018;12:e1700101.
  • Danda R, Ganapathy K, Sathe G, et al. Proteomic profiling of retinoblastoma by high resolution mass spectrometry. Clin Proteomics. 2016;13:29.
  • Naru J, Aggarwal R, Singh U, et al. Proteomic analysis of differentially expressed proteins in vitreous humor of patients with retinoblastoma using iTRAQ-coupled ESI-MS/MS approach. Tumour Biol. 2016;37:13915–13926.
  • Shahulhameed S, Vishwakarma S, Chhablani J, et al. A systematic investigation on complement pathway activation in diabetic retinopathy. Front Immunol [ Internet]. 2020 [cited 2020 Mar 19];11. Available from: https://www.frontiersin.org/articles/10.3389/fimmu.2020.00154/full
  • Parthiban N, Sampath NL, JeyaMaheshwari J, et al. Quantitative profiling of tear proteome reveals down regulation of zinc alpha-2 glycoprotein in Aspergillus flavus keratitis patients. Exp Eye Res. 2019;186:107700.
  • Nikhalashree S, George R, Shantha B, et al. Detection of proteins associated with extracellular matrix regulation in the aqueous humour of patients with primary glaucoma. Curr Eye Res. 2019;44:1018–1025.
  • Basu K, Maurya N, Kaur J, et al. Possible role of differentially expressing novel protein markers (Ligatin and fibulin-7) in human aqueous humor and trabecular meshwork tissue in glaucoma progression. Cell Biol Int. 2019;43:820–834.
  • Goel R, Murthy KR, Srikanth SM, et al. Characterizing the normal proteome of human ciliary body. Clin Proteomics. 2013;10:9.
  • Kaur I, Kaur J, Sooraj K, et al. Comparative evaluation of the aqueous humor proteome of primary angle closure and primary open angle glaucomas and age-related cataract eyes. Int Ophthalmol. 2019;39:69–104.
  • Murthy KR, Rajagopalan P, Pinto SM, et al. Proteomics of human aqueous humor. OMICS. 2015;19:283–293.
  • Karthikkeyan G, Subbannayya Y, Najar MA, et al. Human optic nerve: an enhanced proteomic expression profile. OMICS. 2018;22:642–652.
  • Srivastava SK. Adoption of electronic health records: a roadmap for India. Healthc Inform Res. 2016;22:261–269.
  • Chaturvedi S, Srinivas KR, Muthuswamy V. Biobanking and privacy in India. J Law Med Ethics. 2016;44:45–57.
  • Yadav BK, Bihari C. Biobanking in India and its importance in cancer research. Edorium J Cancer. 2017;3:10–12.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.