1
|
Bhogal I, Pankaj V, Roy S. Identifying RAGE inhibitors as potential therapeutics for Alzheimer's disease via integrated in-silico approaches. Sci Rep 2025; 15:17730. [PMID: 40404684 PMCID: PMC12098779 DOI: 10.1038/s41598-025-01271-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized by two hallmarks: amyloid beta plaques and neurofibrillary tangles. The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor involved in the pathophysiology of various diseases including cancer, diabetes, cardiovascular diseases, and Alzheimer's disease (AD). Therefore, targeting RAGE could be an effective strategy to block RAGE signaling pathways. The present study aims to identify potential RAGE inhibitors against AD through comprehensive in-silico approaches. A total of 708,580 compounds were screened from numerous databases using structure-based virtual screening and ADMET evaluation. Further, the molecules with good glide scores were assessed by molecular docking studies. Subsequently, the top six ligands were subjected to molecular dynamic (MD) simulations for 100 ns and binding free energy calculations to check their stability with RAGE (PDB: 6XQ3). The per-residue decomposition analysis revealed that specific residues namely, GLY_20, ALA_21, LYS_39, GLU_50, LYS_52, ARG_98, GLN_100, LYS_110, ASN_112, and ARG_198 played a key role in the binding process. Furthermore, the trajectory analysis (DCCM and PCA) analyzed the dominant motions of residues and predicted the stability of protein-ligand complexes. In conclusion, the Hit-6 compound could be a promising candidate for targeting RAGE and deserves further consideration as an anti-Alzheimer drug.
Collapse
Affiliation(s)
- Inderjeet Bhogal
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, 616 00, Czech Republic
| | - Vaishali Pankaj
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, 616 00, Czech Republic
| | - Sudeep Roy
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, 616 00, Czech Republic.
| |
Collapse
|
2
|
Chinnathambi S. Histone deacetylase's regulates Tau function in Alzheimer's disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 143:339-361. [PMID: 39843140 DOI: 10.1016/bs.apcsb.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disease associated with dementia and neuronal impairments in brain. AD is characterized histopathologically by two hallmark lesions: abnormally phosphorylated Tau inside neurons as intracellular NFTs and extracellular accumulation of amyloid β peptide (Aβ). Furthermore, it is unable to clarify the distinction between the brief association between the development and build-up of Aβ and the commencement of illness. Additionally, a number of experimental findings suggest that symptoms related to Aβ may only manifest within the framework of anabatic Tauopathies. Tau, a natively unfolded protein, essentially involved in microtubule binding and assembly. Tau protein consists of truncated segment and the purpose of this truncated fragment is to initiate and promote the conversion of soluble Tau into aggregates. The most common aberrant posttranslational change found in Neuro Fibrillary Tangles is hyperphosphorylation, which is essentially composed of aggregated Tau. Tau phosphorylation and acetylation of Tau protein at the locations controlled by histone deacetylase 6 compete, which modulates Tau function. Considering the potential benefits of targeting HDAC6 in AD, we propose focusing on the role of HDAC6 in regulating Tau functions and the other targets are the therapeutic understanding of AD.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India.
| |
Collapse
|
3
|
Miyagawa T, Iwata Y, Oshima M, Ogura H, Sato K, Nakagawa S, Yamamura Y, Kamikawa Y, Miyake T, Kitajima S, Toyama T, Hara A, Sakai N, Shimizu M, Furuichi K, Munesue S, Yamamoto Y, Kaneko S, Wada T. Soluble receptor for advanced glycation end products protects from ischemia- and reperfusion-induced acute kidney injury. Biol Open 2021; 11:273473. [PMID: 34812852 PMCID: PMC8822355 DOI: 10.1242/bio.058852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/11/2021] [Indexed: 11/20/2022] Open
Abstract
The full-length receptor for advanced glycation end products (RAGE) is a multiligand pattern recognition receptor. High-mobility group box 1 (HMGB1) is a RAGE ligand of damage-associated molecular patterns that elicits inflammatory reactions. The shedded isoform of RAGE and endogenous secretory RAGE (esRAGE), a splice variant, are soluble isoforms (sRAGE) that act as organ-protective decoys. However, the pathophysiologic roles of RAGE/sRAGE in acute kidney injury (AKI) remain unclear. We found that AKI was more severe, with enhanced renal tubular damage, macrophage infiltration, and fibrosis, in mice lacking both RAGE and sRAGE than in wild-type control mice. Using murine tubular epithelial cells (TECs), we demonstrated that hypoxia upregulated messenger RNA (mRNA) expression of HMGB1 and tumor necrosis factor α (TNF-α), whereas RAGE and esRAGE expressions were paradoxically decreased. Moreover, the addition of recombinant sRAGE canceled hypoxia-induced inflammation and promoted cell viability in cultured TECs. sRAGE administration prevented renal tubular damage in models of ischemia/reperfusion-induced AKI and of anti-glomerular basement membrane (anti-GBM) glomerulonephritis. These results suggest that sRAGE is a novel therapeutic option for AKI.
Collapse
Affiliation(s)
- Taro Miyagawa
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Yasunori Iwata
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan.,Division of Infection Control, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Megumi Oshima
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Hisayuki Ogura
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Koichi Sato
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Shiori Nakagawa
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Yuta Yamamura
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Yasutaka Kamikawa
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Taito Miyake
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Shinji Kitajima
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Tadashi Toyama
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Akinori Hara
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Norihiko Sakai
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan.,Division of Blood Purification, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Miho Shimizu
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Kengo Furuichi
- Department of Nephrology, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa 920-0293, Japan
| | - Seiichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Shuichi Kaneko
- Department of System Biology, Institute of Medical Pharmaceutical and Health Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan
| |
Collapse
|
4
|
Wu KJ, Wang W, Wang HMD, Leung CH, Ma DL. Interfering with S100B-effector protein interactions for cancer therapy. Drug Discov Today 2020; 25:1754-1761. [PMID: 32679172 DOI: 10.1016/j.drudis.2020.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/17/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022]
Abstract
S100 calcium-binding protein B (S100B) is overexpressed in various malignant tumors, where it regulates cancer cell proliferation and metabolism by physical interactions with other molecules. Interfering with S100B-effector protein interactions is a potential strategy to treat malignant tumors. Although some S100B inhibitors have been discovered by virtual screening (VS), most target the S100B-p53 interaction. Hence, there is scope for the discovery of other S100B-effector protein interaction modulators for malignant tumors. In this review, we provide an overview of S100B-effector protein interaction inhibitor discovery using VS and discuss promising S100B-effector protein interaction targets that permit in silico analysis for drug discovery.
Collapse
Affiliation(s)
- Ke-Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macao SAR, China
| | - Wanhe Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong, China
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macao SAR, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong, China.
| |
Collapse
|
5
|
Dariya B, Nagaraju GP. Advanced glycation end products in diabetes, cancer and phytochemical therapy. Drug Discov Today 2020; 25:1614-1623. [PMID: 32652310 DOI: 10.1016/j.drudis.2020.07.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/04/2020] [Accepted: 07/03/2020] [Indexed: 02/08/2023]
Abstract
The irreversible glycation and oxidation of proteins and lipids produces advanced glycation end products (AGEs). These modified AGEs are triggered to bind the receptor for AGE (RAGE), thereby activating its downstream signaling pathways, such as nuclear factor (NF)-κB and phosphoinositide 3-kinase (PI3K)/Akt, ultimately leading to diabetes and cancers. In this review, we focus on the interaction of AGE-RAGE and their associated pathways. We also consider the activity of phytochemicals, such as genistein and curcumin, that trap dicarbonyl compounds including methylglyoxal (MG) and glyoxalase that arise from multiple pathways to block AGE formation and prevent its interaction with RAGE.
Collapse
Affiliation(s)
- Begum Dariya
- Department of Biosciences and Biotechnology, Banasthali University, Banasthali, Rajasthan 304022, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
6
|
Watanabe M, Toyomura T, Wake H, Liu K, Teshigawara K, Takahashi H, Nishibori M, Mori S. Differential contribution of possible pattern-recognition receptors to advanced glycation end product-induced cellular responses in macrophage-like RAW264.7 cells. Biotechnol Appl Biochem 2019; 67:265-272. [PMID: 31654427 DOI: 10.1002/bab.1843] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/19/2019] [Indexed: 01/10/2023]
Abstract
Advanced glycation end products (AGEs) are considered to be related to the pathogenesis of some inflammatory diseases. AGEs were reported to stimulate the receptor for AGEs (RAGE), which causes inflammatory reactions. However, recently, toll-like receptors (TLRs), in addition to RAGE, have been reported to be related to AGE-mediated cellular responses, and it remains unclear which receptor is responsible for AGE recognition. To reveal the role of pattern-recognition receptors, including TLRs and/or RAGE, in AGE-mediated cellular responses, we generated macrophage-like RAW264.7 knockout (KO) cells lacking these receptors by genome editing using the CRISPR/Cas9 system and assessed AGE-stimulated changes in these cells. Comparison of the established clones suggested that RAGE partially affects the expression of TLRs. In the KO clone lacking TLR4 and TLR2, AGE-stimulated tumor necrosis factor alpha (TNF-α) expression and phosphorylation of IκBα, p38, and extracellular signal-regulated kinase (ERK) were significantly attenuated, suggesting that AGE-mediated responses are largely dependent on TLRs. On the other hand, on comparison of the AGE-stimulated responses between the KO clone lacking TLR4 and TLR2, and the clone lacking TLR4, TLR2, and RAGE, RAGE played little role in AGE-stimulated TNF-α transcription and ERK phosphorylation. Taken together, this study suggested that AGE-stimulated inflammatory responses occur mainly through TLRs rather than RAGE.
Collapse
Affiliation(s)
- Masahiro Watanabe
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Takao Toyomura
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Hidenori Wake
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Keyue Liu
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Kiyoshi Teshigawara
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hideo Takahashi
- Department of Pharmacology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Shuji Mori
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| |
Collapse
|
7
|
Farhadi A, Vosough M, Zhang JS, Tahamtani Y, Shahpasand K. A Possible Neurodegeneration Mechanism Triggered by Diabetes. Trends Endocrinol Metab 2019; 30:692-700. [PMID: 31399291 DOI: 10.1016/j.tem.2019.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/08/2019] [Accepted: 07/12/2019] [Indexed: 01/14/2023]
Abstract
Several conditions result in neurodegeneration; among which diabetes mellitus (DM) is of crucial importance. Tau (τ) malfunction is a major pathological process participating in neurodegeneration. Despite extensive considerations, the actual causative link between DM and τ abnormalities remains uncertain thus far. Phosphorylated (p)-τ at Thr-Pro motifs can exist in the two distinct cis and trans conformations. cis is neurotoxic, and is accumulated upon various stress conditions, such as nutrition depletion. We assume that pathogenic cis p-τ is the central mediator of neurodegeneration in DM, and propose why different brain areas give various responses to stress conditions. We herein juxtapose recent approaches in diabetic neurodegeneration and propose a therapeutic target to stop neuronal loss during DM.
Collapse
Affiliation(s)
- Aisan Farhadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, IR 19395-4644, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Jin-San Zhang
- International Collaborative Center on Growth Factor Research, and School of Pharmaceutical Sciences, Wenzhou Medical University; Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China; Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, IR 19395-4644, Iran.
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, IR 19395-4644, Iran.
| |
Collapse
|
8
|
Rathore R, Sonwane BP, Jagadeeshaprasad MG, Kahar S, Santhakumari B, Unnikrishnan AG, Kulkarni MJ. Glycation of glucose sensitive lysine residues K36, K438 and K549 of albumin is associated with prediabetes. J Proteomics 2019; 208:103481. [PMID: 31394310 DOI: 10.1016/j.jprot.2019.103481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 12/16/2022]
Abstract
Prediabetes is a risk factor for the development of diabetes. Early diagnosis of prediabetes may prevent the onset and progression of diabetes and its associated complications. Therefore, this study aimed at the identification of novel markers for efficient prediction of prediabetes. In this pursuit, we have evaluated the ability of glycated peptides of albumin in predicting prediabetes. Glycated peptides of in vitro glycated albumin were characterized by data dependent acquisition and parallel reaction monitoring using LC-HRMS. Amongst 14 glycated peptides characterized in vitro, four peptides, particularly, FK(CML)DLGEENFK, K(AML)VPQVSTPTLVEVSR, K(CML)VPQVSTPTLVEVSR, and K(AML)QTALVELVK, corresponding to 3 glucose sensitive lysine residues K36, K438, and K549, respectively showed significantly higher abundance in prediabetes than control. Additionally, the abundance of three of these peptides, namely K(AML)QTALVELVK, K(CML)VPQVSTPTLVEVSR and FK(CML)DLGEENFK was >1.8-fold in prediabetes, which was significantly higher than the differences observed for FBG, PPG, and HbA1c. Further, the four glycated peptides showed a significant correlation with FBG, PPG, HbA1c, triglycerides, VLDL, and HDL. This study supports that glycated peptides of glucose sensitive lysine residues K36, K438 and K549 of albumin could be potentially useful markers for prediction of prediabetes. SIGNIFICANCE: Undiagnosed prediabetes may lead to diabetes and associated complications. This study reports targeted quantification of four glycated peptides particulary FK(CML)DLGEENFK, K(AML)VPQVSTPTLVEVSR, K(CML)VPQVSTPTLVEVSR, and K(AML)QTALVELVK, corresponding to 3 glucose sensitive lysine residues K36, K438 and K549 respectively by parallel reaction monitoring in healthy and prediabetic subjects. These peptides showed significantly higher abundance in prediabetes than healthy subjects, and showed significant correlation with various clinical parameters including FBG, PPG, HbA1c, and altered lipid profile. Therefore, together these four peptides constitute a panel of markers that can be useful for prediction of prediabetes.
Collapse
Affiliation(s)
- Rajeshwari Rathore
- Proteomics Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Babasaheb P Sonwane
- Proteomics Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - M G Jagadeeshaprasad
- Proteomics Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | - B Santhakumari
- Proteomics Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | - Mahesh J Kulkarni
- Proteomics Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
9
|
Prion-Like Propagation of Post-Translationally Modified Tau in Alzheimer’s Disease: A Hypothesis. J Mol Neurosci 2018; 65:480-490. [DOI: 10.1007/s12031-018-1111-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/20/2018] [Indexed: 12/25/2022]
|
10
|
Batkulwar K, Godbole R, Banarjee R, Kassaar O, Williams RJ, Kulkarni MJ. Advanced Glycation End Products Modulate Amyloidogenic APP Processing and Tau Phosphorylation: A Mechanistic Link between Glycation and the Development of Alzheimer's Disease. ACS Chem Neurosci 2018; 9:988-1000. [PMID: 29384651 DOI: 10.1021/acschemneuro.7b00410] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Advanced glycation end products (AGEs) are implicated in the pathology of Alzheimer's disease (AD), as they induce neurodegeneration following interaction with the receptor for AGE (RAGE). This study aimed to establish a mechanistic link between AGE-RAGE signaling and AD pathology. AGE-induced changes in the neuro2a proteome were monitored by SWATH-MS. Western blotting and cell-based reporter assays were used to investigate AGE-RAGE regulated APP processing and tau phosphorylation in primary cortical neurons. Selected protein expression was validated in brain samples affected by AD. The AGE-RAGE axis altered proteome included increased expression of cathepsin B and asparagine endopeptidase (AEP), which mediated an increase in Aβ1-42 formation and tau phosphorylation, respectively. Elevated cathepsin B, AEP, RAGE, and pTau levels were found in human AD brain, coincident with enhanced AGEs. This study demonstrates that the AGE-RAGE axis regulates Aβ1-42 formation and tau phosphorylation via increased cathepsin B and AEP, providing a new molecular link between AGEs and AD pathology.
Collapse
Affiliation(s)
- Kedar Batkulwar
- Proteomics Facility, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune-411008, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-National Chemical Laboratory, Pune-411008, India
| | - Rashmi Godbole
- Proteomics Facility, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune-411008, India
| | - Reema Banarjee
- Proteomics Facility, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune-411008, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-National Chemical Laboratory, Pune-411008, India
| | - Omar Kassaar
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, U.K
| | - Robert J. Williams
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, U.K
| | - Mahesh J. Kulkarni
- Proteomics Facility, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune-411008, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-National Chemical Laboratory, Pune-411008, India
| |
Collapse
|
11
|
Jana AK, Batkulwar KB, Kulkarni MJ, Sengupta N. Glycation induces conformational changes in the amyloid-β peptide and enhances its aggregation propensity: molecular insights. Phys Chem Chem Phys 2018; 18:31446-31458. [PMID: 27827482 DOI: 10.1039/c6cp05041g] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The cytotoxicity of the amyloid beta (Aβ) peptide, implicated in the pathogenesis of Alzheimer's disease (AD), can be enhanced by its post-translational glycation, a series of non-enzymatic reactions with reducing sugars and reactive dicarbonyls. However, little is known about the underlying mechanisms that potentially enhance the cytotoxicity of the advanced glycation modified Aβ. In this work, fully atomistic molecular dynamics (MD) simulations are exploited to obtain direct molecular insights into the process of early Aβ self-assembly in the presence and absence of glycated lysine residues. Analyses of data exceeding cumulative timescales of 1 microsecond for each system reveal that glycation results in a stronger enthalpy of association between Aβ monomers and lower conformational entropy, in addition to a sharp overall increase in the beta-sheet content. Further analyses reveal that the enhanced interactions originate, in large part, due to markedly stronger, as well as new, inter-monomer salt bridging propensities in the glycated variety. Interestingly, these conformational and energetic effects are broadly reflected in preformed protofibrillar forms of Aβ small oligomers modified with glycation. Our combined results imply that glycation consolidates Aβ self-assembly regardless of its point of occurrence in the pathway. They provide a basis for further mechanistic studies and therapeutic endeavors that could potentially result in novel ways of combating AGE related AD progression.
Collapse
Affiliation(s)
- Asis K Jana
- Physical Chemistry Division, CSIR-National Chemical Laboratory, Pune 411008, India and Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Kedar B Batkulwar
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India and Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India.
| | - Mahesh J Kulkarni
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India and Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India.
| | - Neelanjana Sengupta
- Dept. of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur 741 246, W. Bengal, India.
| |
Collapse
|
12
|
Gasparotto J, Girardi CS, Somensi N, Ribeiro CT, Moreira JCF, Michels M, Sonai B, Rocha M, Steckert AV, Barichello T, Quevedo J, Dal-Pizzol F, Gelain DP. Receptor for advanced glycation end products mediates sepsis-triggered amyloid-β accumulation, Tau phosphorylation, and cognitive impairment. J Biol Chem 2017; 293:226-244. [PMID: 29127203 DOI: 10.1074/jbc.m117.786756] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 11/09/2017] [Indexed: 11/06/2022] Open
Abstract
Patients recovering from sepsis have higher rates of CNS morbidities associated with long-lasting impairment of cognitive functions, including neurodegenerative diseases. However, the molecular etiology of these sepsis-induced impairments is unclear. Here, we investigated the role of the receptor for advanced glycation end products (RAGE) in neuroinflammation, neurodegeneration-associated changes, and cognitive dysfunction arising after sepsis recovery. Adult Wistar rats underwent cecal ligation and perforation (CLP), and serum and brain (hippocampus and prefrontal cortex) samples were obtained at days 1, 15, and 30 after the CLP. We examined these samples for systemic and brain inflammation; amyloid-β peptide (Aβ) and Ser-202-phosphorylated Tau (p-TauSer-202) levels; and RAGE, RAGE ligands, and RAGE intracellular signaling. Serum markers associated with the acute proinflammatory phase of sepsis (TNFα, IL-1β, and IL-6) rapidly increased and then progressively decreased during the 30-day period post-CLP, concomitant with a progressive increase in RAGE ligands (S100B, Nϵ-[carboxymethyl]lysine, HSP70, and HMGB1). In the brain, levels of RAGE and Toll-like receptor 4, glial fibrillary acidic protein and neuronal nitric-oxide synthase, and Aβ and p-TauSer-202 also increased during that time. Of note, intracerebral injection of RAGE antibody into the hippocampus at days 15, 17, and 19 post-CLP reduced Aβ and p-TauSer-202 accumulation, Akt/mechanistic target of rapamycin signaling, levels of ionized calcium-binding adapter molecule 1 and glial fibrillary acidic protein, and behavioral deficits associated with cognitive decline. These results indicate that brain RAGE is an essential factor in the pathogenesis of neurological disorders following acute systemic inflammation.
Collapse
Affiliation(s)
- Juciano Gasparotto
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003 RS, Brazil
| | - Carolina S Girardi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003 RS, Brazil
| | - Nauana Somensi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003 RS, Brazil
| | - Camila T Ribeiro
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003 RS, Brazil
| | - José C F Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003 RS, Brazil
| | - Monique Michels
- Laboratório de Fisiopatologia Experimental, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Criciúma 88806-000 SC, Brazil
| | - Beatriz Sonai
- Laboratório de Fisiopatologia Experimental, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Criciúma 88806-000 SC, Brazil
| | - Mariane Rocha
- Laboratório de Fisiopatologia Experimental, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Criciúma 88806-000 SC, Brazil
| | - Amanda V Steckert
- Laboratório de Neurociências at Programa de Pós-GraduaçΔo em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense-Criciúma, Criciúma 88806-000 SC, Brazil; Translational Psychiatry Program, University of Texas Health Science Center at Houston, Houston, Texas 77030; Center of Excellence on Mood Disorders at Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030; Neuroscience Graduate Program, University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030
| | - Tatiana Barichello
- Laboratório de Neurociências at Programa de Pós-GraduaçΔo em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense-Criciúma, Criciúma 88806-000 SC, Brazil; Translational Psychiatry Program, University of Texas Health Science Center at Houston, Houston, Texas 77030; Center of Excellence on Mood Disorders at Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030; Neuroscience Graduate Program, University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030
| | - JoΔo Quevedo
- Laboratório de Neurociências at Programa de Pós-GraduaçΔo em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense-Criciúma, Criciúma 88806-000 SC, Brazil; Translational Psychiatry Program, University of Texas Health Science Center at Houston, Houston, Texas 77030; Center of Excellence on Mood Disorders at Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030; Neuroscience Graduate Program, University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030
| | - Felipe Dal-Pizzol
- Laboratório de Fisiopatologia Experimental, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Criciúma 88806-000 SC, Brazil
| | - Daniel P Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003 RS, Brazil.
| |
Collapse
|
13
|
Matsumoto H, Matsumoto N, Shimazaki J, Nakagawa J, Imamura Y, Yamakawa K, Yamada T, Ikeda M, Hiraike H, Ogura H, Shimazu T. Therapeutic Effectiveness of Anti-RAGE Antibody Administration in a Rat Model of Crush Injury. Sci Rep 2017; 7:12255. [PMID: 28947744 PMCID: PMC5613014 DOI: 10.1038/s41598-017-12065-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 09/04/2017] [Indexed: 11/09/2022] Open
Abstract
Crush injury patients often have systemic inflammatory response syndrome that leads to multiple organ failure. Receptor for advanced glycation endproducts (RAGE) functions as a pattern recognition receptor that regulates inflammation. We evaluated the effects of anti-RAGE antibody in a crush injury model. Pressure was applied to both hindlimbs of rats for 6 h by 3.0-kg blocks and then released. Animals were randomly divided into the sham (RAGE-Sh) group, crush (RAGE-Ctrl) group or anti-RAGE antibody-treated crush (RAGE-Tx) group. Samples were collected at 3, 6 and 24 h after releasing pressure. In the RAGE-Ctrl group, fluorescent immunostaining in the lung showed upregulated RAGE expression at 3 h. The serum soluble RAGE (sRAGE) level, which reflects the amount of RAGE expression in systemic tissue, increased at 6 h. Serum interleukin 6 (IL-6; systemic inflammation marker) increased immediately at 3 h. Histological analysis revealed lung injury at 6 and 24 h. Administration of anti-RAGE antibody before releasing compression inhibited upregulated RAGE expression in the lung alveoli, suppressed RAGE-associated mediators sRAGE and IL6, attenuated the lung damage and improved the 7-day survival rate. Collectively, our results indicated that the use of anti-RAGE antibody before releasing compression is associated with a favourable prognosis following crush injury.
Collapse
Affiliation(s)
- Hisatake Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan.
| | - Naoya Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Junya Shimazaki
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Junichiro Nakagawa
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Yukio Imamura
- Laboratory of Nano-Bio Probes, RIKEN Quantitative Biology Center, 6-2-3 Furuedai, Suita, Osaka, 565-0874, Japan
| | - Kazuma Yamakawa
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Tomoki Yamada
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Mitsunori Ikeda
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Hiroko Hiraike
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka Suita, Osaka, 565-0871, Japan
| |
Collapse
|
14
|
Xie Y, Yu N, Chen Y, Zhang K, Ma HY, Di Q. HMGB1 regulates P-glycoprotein expression in status epilepticus rat brains via the RAGE/NF-κB signaling pathway. Mol Med Rep 2017. [PMID: 28627626 PMCID: PMC5562060 DOI: 10.3892/mmr.2017.6772] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Overexpression of P-glycoprotein (P-gp) in the brain is an important mechanism involved in drug-resistant epilepsy (DRE). High-mobility group box 1 (HMGB1), an inflammatory cytokine, significantly increases following seizures and may be involved in upregulation of P-gp. However, the underlying mechanisms remain elusive. The aim of the present study was to evaluate the role of HMGB1 and its downstream signaling components, receptor for advanced glycation end-product (RAGE) and nuclear factor-κB (NF-κB), on P-gp expression in rat brains during status epilepticus (SE). Small interfering RNA (siRNA) was administered to rats prior to induction of SE by pilocarpine, to block transcription of the genes encoding HMGB1 and RAGE, respectively. An inhibitor of NF-κB, pyrrolidinedithiocarbamic acid (PDTC), was utilized to inhibit activation of NF-κB. The expression levels of HMGB1, RAGE, phosphorylated-NF-κB p65 (p-p65) and P-gp were detected by western blotting. The relative mRNA expression levels of the genes encoding these proteins were measured using reverse transcription-quantitative polymerase chain reaction and the cellular localization of the proteins was determined by immunofluorescence. Pre-treatment with HMGB1 siRNA reduced the expression levels of RAGE, p-p65 and P-gp. PDTC reduced the expression levels of P-gp. These findings suggested that overexpression of P-gp during seizures may be regulated by HMGB1 via the RAGE/NF-κB signaling pathway, and may be a novel target for treating DRE.
Collapse
Affiliation(s)
- Yuan Xie
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Nian Yu
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Chen
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Kang Zhang
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hai-Yan Ma
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qing Di
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
15
|
Hormetic and regulatory effects of lipid peroxidation mediators in pancreatic beta cells. Mol Aspects Med 2016; 49:49-77. [PMID: 27012748 DOI: 10.1016/j.mam.2016.03.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/23/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022]
Abstract
Nutrient sensing mechanisms of carbohydrates, amino acids and lipids operate distinct pathways that are essential for the adaptation to varying metabolic conditions. The role of nutrient-induced biosynthesis of hormones is paramount for attaining metabolic homeostasis in the organism. Nutrient overload attenuate key metabolic cellular functions and interfere with hormonal-regulated inter- and intra-organ communication, which may ultimately lead to metabolic derangements. Hyperglycemia and high levels of saturated free fatty acids induce excessive production of oxygen free radicals in tissues and cells. This phenomenon, which is accentuated in both type-1 and type-2 diabetic patients, has been associated with the development of impaired glucose tolerance and the etiology of peripheral complications. However, low levels of the same free radicals also induce hormetic responses that protect cells against deleterious effects of the same radicals. Of interest is the role of hydroxyl radicals in initiating peroxidation of polyunsaturated fatty acids (PUFA) and generation of α,β-unsaturated reactive 4-hydroxyalkenals that avidly form covalent adducts with nucleophilic moieties in proteins, phospholipids and nucleic acids. Numerous studies have linked the lipid peroxidation product 4-hydroxy-2E-nonenal (4-HNE) to different pathological and cytotoxic processes. Similarly, two other members of the family, 4-hydroxyl-2E-hexenal (4-HHE) and 4-hydroxy-2E,6Z-dodecadienal (4-HDDE), have also been identified as potential cytotoxic agents. It has been suggested that 4-HNE-induced modifications in macromolecules in cells may alter their cellular functions and modify signaling properties. Yet, it has also been acknowledged that these bioactive aldehydes also function as signaling molecules that directly modify cell functions in a hormetic fashion to enable cells adapt to various stressful stimuli. Recent studies have shown that 4-HNE and 4-HDDE, which activate peroxisome proliferator-activated receptor δ (PPARδ) in vascular endothelial cells and insulin secreting beta cells, promote such adaptive responses to ameliorate detrimental effects of high glucose and diabetes-like conditions. In addition, due to the electrophilic nature of these reactive aldehydes they form covalent adducts with electronegative moieties in proteins, phosphatidylethanolamine and nucleotides. Normally these non-enzymatic modifications are maintained below the cytotoxic range due to efficient cellular neutralization processes of 4-hydroxyalkenals. The major neutralizing enzymes include fatty aldehyde dehydrogenase (FALDH), aldose reductase (AR) and alcohol dehydrogenase (ADH), which transform the aldehyde to the corresponding carboxylic acid or alcohols, respectively, or by biding to the thiol group in glutathione (GSH) by the action of glutathione-S-transferase (GST). This review describes the hormetic and cytotoxic roles of oxygen free radicals and 4-hydroxyalkenals in beta cells exposed to nutritional challenges and the cellular mechanisms they employ to maintain their level at functional range below the cytotoxic threshold.
Collapse
|
16
|
Lin JA, Wu CH, Lu CC, Hsia SM, Yen GC. Glycative stress from advanced glycation end products (AGEs) and dicarbonyls: An emerging biological factor in cancer onset and progression. Mol Nutr Food Res 2016; 60:1850-64. [DOI: 10.1002/mnfr.201500759] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/11/2015] [Accepted: 01/07/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Jer-An Lin
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
| | - Chi-Hao Wu
- School of Nutrition and Health Sciences; Taipei Medical University; Taipei Taiwan
| | - Chi-Cheng Lu
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
- School of Nutrition and Health Sciences; Taipei Medical University; Taipei Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences; Taipei Medical University; Taipei Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
- Agricultural Biotechnology Center; National Chung Hsing University; Taichung Taiwan
| |
Collapse
|
17
|
Kolekar YM, Vannuruswamy G, Bansode SB, B S, Thulasiram HV, Kulkarni MJ. Investigation of antiglycation activity of isoprenaline. RSC Adv 2015. [DOI: 10.1039/c5ra01723h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Isoprenaline reduces the advanced glycation end products and may be suitable candidate for the treatment of glycation associated diseases.
Collapse
Affiliation(s)
- Yogesh M. Kolekar
- Proteomics Facility
- Division of Biochemical Sciences
- CSIR-National Chemical Laboratory
- Pune-411008
- India
| | - Garikapati Vannuruswamy
- Proteomics Facility
- Division of Biochemical Sciences
- CSIR-National Chemical Laboratory
- Pune-411008
- India
| | - Sneha B. Bansode
- Proteomics Facility
- Division of Biochemical Sciences
- CSIR-National Chemical Laboratory
- Pune-411008
- India
| | - Santhakumari B
- Centre for Material Characterization
- CSIR-National Chemical Laboratory
- Pune-411008
- India
| | | | - Mahesh J. Kulkarni
- Proteomics Facility
- Division of Biochemical Sciences
- CSIR-National Chemical Laboratory
- Pune-411008
- India
| |
Collapse
|