1
|
Wang B, Liu S, Hao K, Wang Y, Li Z, Lou Y, Chang Y, Qi W. HDAC6 modulates the cognitive behavioral function and hippocampal tissue pathological changes of APP/PS1 transgenic mice through HSP90-HSF1 pathway. Exp Brain Res 2024; 242:1983-1998. [PMID: 38935089 DOI: 10.1007/s00221-024-06858-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024]
Abstract
The aim of this study was to investigate histone deacetylase 6 (HDAC6) modifies the heat shock protein 90 (HSP90) and heat shock transcription factor 1 (HSF1) affect the levels of pathological markers such as Aβ oligomers (Aβo) and Tau phosphorylation (p-Tau) in APP/PS1 double transgenic mice hippocampal tissues or HT22 neurons as well as the changes in cognitive behavioral functions of mice. (1) APP/PS1 transgenic mice (6 months old, 25 ~ 30 g) were randomly assigned to 5 experimental groups, C57BL/6J mice (6 months old, 25 ~ 30 g) were used as 4 control groups, with 8 mice in each group. All mice underwent intracerebroventricular (i.c.v.) cannulation, and the experimental groups were administered with normal saline (APP + NS group), HDAC6 agonist tubastatin A hydrochloride (TSA) (APP + TSA group) or HDAC6 agonist theophylline (Theo) (APP + Theo group), HSP90 inhibitor Ganetespib (Gane) (APP + Gane group), or a combination of pre-injected Gane by TSA (APP + Gane + TSA group); the control group received i.c.v. injections of Gane (Gane group), TSA (TSA group), Theo (Theo group) or NS (NS group), respectively. (2) Mouse hippocampal neurons HT22 were randomly divided into a control group (Control) and an Aβ1-42 intervention group (Aβ). Within the Aβ group, further divisions were made for knockdown HSP90 (Aβ + siHSP90 group), overexpression HSP90 (Aβ + OE-HSP90 group), knockdown HSF1(Aβ + siHSF1 group) and knockdown HSF1 followed by overexpression HSP90 (Aβ + siHSF1 + OE-HSP90 group), resulting in a total of 6 groups. Morris water maze test was used to evaluate the cognitive behavior of the mice. Western blot and immunohistochemistry or immunofluorescence were performed to detect the levels of HDAC6, HSP90, HSF1, Aβ1-42, Tau protein, and p-Tau in the hippocampal tissue or HT22 cells. qRT-PCR was used to measure the levels of hdac6, hsp90, and hsf1 mRNA in the hippocampus or nerve cells. (1) The levels of HDAC6, Aβ1-42 and p-Tau were elevated, while HSP90 and HSF1 were decreased in the hippocampal tissue of APP/PS1 transgenic mice (all P < 0.01). Inhibiting HDAC6 upregulated the expressions of HSP90 and HSF1 in the hippocampal tissue of APP/PS1 mice, while decreasing the levels of Aβ1-42 and p-Tau as well as improving the spatial cognitive behavior in mice (P < 0.05 or P < 0.01). The opposite effects were observed upon HDAC6 activation. However, inhibiting HSP90 reduced the expression of HSF1 (P < 0.01) and increased the levels of Aβ1-42 and p-Tau (P < 0.05 or P < 0.01) but did not significantly affect the expression of HDAC6 (P > 0.05). No significant changes were observed in the aforementioned indicators in the 4 control groups (P > 0.05). (2) In the Aβ1-42 intervention group, HDAC6 and Aβ1-42, p-Tau expression levels were elevated, while HSP90 and HSF1 expressions were all decreased, and cell viability was reduced (P < 0.05 or P < 0.01). Overexpression of HSP90 upregulated HSF1 expression, decreased the levels of Aβ1-42 and p-Tau, and increased cell viability (P < 0.05 or P < 0.01). Knocking down HSP90 had the opposite effect; and knocking down HSF1 increased the levels of Aβ1-42 and p-Tau and decreased cells viability (all P < 0.01), but did not result in significant changes in the expression levels of HSP90 (P > 0.05). Inhibiting HDAC6 can upregulate the expressions of HSP90 and HSF1 but reduce the levels of Aβ1-42 and p-Tau in the hippocampus of APP/PS1 mice and improvement of cognitive behavioral function in mice; Overexpression of HSP90 can increase HSF1 but decrease Aβ1-42 and p-Tau levels in the hippocampal neurons and increase cell activity. It is suggested that HDAC6 may affect the formation of Aβ oligomers and the changes in Tau protein phosphorylation levels in the hippocampus of AD transgenic mouse as well as the alterations in cognitive behavioral functions by regulating the HSP90-HSF1 pathway.
Collapse
Affiliation(s)
- Bingyi Wang
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, China
| | - Siyu Liu
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, China
| | - Kaimin Hao
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, China
| | - YaruWang Wang
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, China
| | - Zongjing Li
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, China
| | - Yuanyuan Lou
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, China
| | - Yuan Chang
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, China
| | - Wenxiu Qi
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, China.
| |
Collapse
|
2
|
Gohda K. Conformational Analysis of the Loop-to-Helix Transition of the α-Helix3 Plastic Region in the N-Terminal Domain of Human Hsp90α by a Computational Biochemistry Approach. J Chem Inf Model 2022; 62:5699-5714. [PMID: 36278922 DOI: 10.1021/acs.jcim.2c00984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hsp90 is a chaperone protein aiding in correct protein folding and attractive for drug discovery. The structure of human Hsp90α N-terminal domain (NTD) is intriguing since the α-helix3 region of the ATP-binding site in the NTD plastically changes its conformation, i.e., loop-out, loop-in, and helical conformations, according to the bound inhibitor type. The plastic region structure is known to influence the mode of inhibition-inhibitors bound to a helix have a longer residence time in the complex, which is a factor of in vivo-active drugs, compared with loop binders. In this study, we analyzed the loop-to-helix transition of the plastic region through binding of a helix binder by a computational biochemistry approach. To generate the helical transition from the loop, the resorcinol inhibitor C1 complexed with a loop-in structure was alchemically transformed to the C10 inhibitor, which is known as a helix binder. The loop in the C1 complex possesses Leu107 tightly binding to the hydrophobic subpocket, considered as a key residue for the plasticity. From 10 × 1 μs simulations after the alchemical transformation, the helical transition was observed with a 29% success rate. Conformational analysis of the simulations identified residues possibly associated with the helical transition. The implementation of additional simulations (dihedral-constrained and in silico mutant simulations) led to a statistically significant increase in the transition success rate to 78%, as observed in Asn105 psi-constrained simulation. Therefore, we concluded that the Asn105 psi dihedral angle is most likely involved in the helical transition by a change of the dihedral angle to gauche-negative.
Collapse
Affiliation(s)
- Keigo Gohda
- Computer-aided Molecular Modeling Research Center, Kansai (CAMM-Kansai), 3-32-302, Tsuto-Otsuka, Nishinomiya 663-8241, Japan
| |
Collapse
|
3
|
Wu Y, Shen S, Shi Y, Tian N, Zhou Y, Zhang X. Senolytics: Eliminating Senescent Cells and Alleviating Intervertebral Disc Degeneration. Front Bioeng Biotechnol 2022; 10:823945. [PMID: 35309994 PMCID: PMC8924288 DOI: 10.3389/fbioe.2022.823945] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/21/2022] [Indexed: 12/25/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is the main cause of cervical and lumbar spondylosis. Over the past few years, the relevance between cellular senescence and IVDD has been widely studied, and the senescence-associated secretory phenotype (SASP) produced by senescent cells is found to remodel extracellular matrix (ECM) metabolism and destruct homeostasis. Elimination of senescent cells by senolytics and suppression of SASP production by senomorphics/senostatics are effective strategies to alleviate degenerative diseases including IVDD. Here, we review the involvement of senescence in the process of IVDD; we also discuss the potential of senolytics on eliminating senescent disc cells and alleviating IVDD; finally, we provide a table listing senolytic drugs and small molecules, aiming to propose potential drugs for IVDD therapy in the future.
Collapse
Affiliation(s)
- Yuhao Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shiwei Shen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| |
Collapse
|
4
|
Larghi EL, Bruneau A, Sauvage F, Alami M, Vergnaud-Gauduchon J, Messaoudi S. Synthesis and Biological Activity of 3-(Heteroaryl)quinolin-2(1 H)-ones Bis-Heterocycles as Potential Inhibitors of the Protein Folding Machinery Hsp90. Molecules 2022; 27:412. [PMID: 35056725 PMCID: PMC8778022 DOI: 10.3390/molecules27020412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 01/02/2023] Open
Abstract
In the context of our SAR study concerning 6BrCaQ analogues as C-terminal Hsp90 inhibitors, we designed and synthesized a novel series of 3-(heteroaryl)quinolin-2(1H), of types 3, 4, and 5, as a novel class of analogues. A Pd-catalyzed Liebeskind-Srogl cross-coupling was developed as a convenient approach for easy access to complex purine architectures. This series of analogues showed a promising biological effect against MDA-MB231 and PC-3 cancer cell lines. This study led to the identification of the best compounds, 3b (IC50 = 28 µM) and 4e, which induce a significant decrease of CDK-1 client protein and stabilize the levels of Hsp90 and Hsp70 without triggering the HSR response.
Collapse
Affiliation(s)
- Enrique L. Larghi
- CNRS, BioCIS, Université Paris-Saclay, 92290 Châtenay-Malabry, France;
- Instituto de Química Rosario (IQUIR) CONICET/UNR, FBioyF, Rosario S2002LRK, Argentina;
| | - Alexandre Bruneau
- Instituto de Química Rosario (IQUIR) CONICET/UNR, FBioyF, Rosario S2002LRK, Argentina;
| | - Félix Sauvage
- CNRS, Institut Galien-Paris Saclay, Université Paris-Saclay, 92296 Châtenay-Malabry, France; (F.S.); (J.V.-G.)
| | - Mouad Alami
- CNRS, BioCIS, Université Paris-Saclay, 92290 Châtenay-Malabry, France;
| | - Juliette Vergnaud-Gauduchon
- CNRS, Institut Galien-Paris Saclay, Université Paris-Saclay, 92296 Châtenay-Malabry, France; (F.S.); (J.V.-G.)
| | - Samir Messaoudi
- CNRS, BioCIS, Université Paris-Saclay, 92290 Châtenay-Malabry, France;
| |
Collapse
|
5
|
Park EJ, Jin SW, Lim HJ, Kim HY, Kang MS, Yang S. Whole Cigarette Smoke Condensates Induce Accumulation of Amyloid Beta Precursor Protein with Oxidative Stress in Murine Astrocytes. TOXICS 2021; 9:150. [PMID: 34203397 PMCID: PMC8309752 DOI: 10.3390/toxics9070150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 11/16/2022]
Abstract
Although cigarette smoking has been postulated to be a potential risk factor for Alzheimer's disease (AD), the toxic mechanism is still unclear. Additionally, astrocytes have been identified as a potential target, given they play multiple roles in maintaining normal brain function. In this study, we explored the toxic mechanism of whole cigarette smoke condensates (WCSC) using murine astrocytes. Cell proliferation, the percentage of cells in the G2/M phase, and LDH concentrations in the cell supernatants were all reduced in WCSC-treated cells. In addition, oxidative stress was induced, together with shortening of processes, structural damage of organelles, disturbances in mitochondrial function, blockage of autophagic signals, accumulation of amyloid β precursor protein, and loss of chemotactic functions. Based on these results, we hypothesize that dysfunction of astrocytes may contribute to the occurrence of cigarette-smoking-induced AD.
Collapse
Affiliation(s)
- Eun-Jung Park
- East–West Medical Science Research Institute, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul 02447, Korea
- Human Health and Environmental Toxins Research Center, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul 02447, Korea
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.-W.J.); (H.-J.L.); (M.-S.K.)
| | - Seung-Woo Jin
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.-W.J.); (H.-J.L.); (M.-S.K.)
| | - Hyun-Ji Lim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.-W.J.); (H.-J.L.); (M.-S.K.)
| | - Hyeon-Young Kim
- Inhalation Toxicology Center for Airborne Risk Factors, Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup 56212, Korea;
| | - Min-Sung Kang
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.-W.J.); (H.-J.L.); (M.-S.K.)
- General Toxicology & Research Group, Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup 56212, Korea
| | - Siyoung Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea;
- Degenerative InterDiseases Research Center, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
6
|
The Prognostic Significance of Hsp70 in Patients with Colorectal Cancer Patients: A PRISMA-Compliant Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5526327. [PMID: 33954173 PMCID: PMC8064787 DOI: 10.1155/2021/5526327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022]
Abstract
Background Hsp70 (heat shock protein 70) plays a key role in carcinogenesis and cancer progression. However, the relationship between the Hsp70 expression level and the colorectal cancer patient survival is unknown. This study is aimed at investigating the relationship between Hsp70 and the prognosis of colorectal carcinoma patients. Methods PubMed, Web of Science, and Embase were used for systematic computer literature retrieval. Stata SE14.0 software was used for quantitative meta-analysis. Besides, data was extracted from selected articles. Relationships between Hsp70 expression level and prognosis were further studied. The hazard ratios (HRs) and 95% confidence intervals (95% CIs) were also computed. Results A total of 11 potentially eligible studies with 2269 patients were identified in 10 tumors from PubMed, Web of Science, and Embase. Hsp70 overexpression was associated with poor overall survival (OS) and disease-free survival (DFS) in colorectal carcinoma patients (HRs, 0.65 (95% CI: 0.52-0.78) and 0.77 (95% CI: 0.23-1.32), respectively). Conclusions Hsp70 overexpression can predict poor survival in colorectal cancer patients.
Collapse
|
7
|
Design of Disruptors of the Hsp90-Cdc37 Interface. Molecules 2020; 25:molecules25020360. [PMID: 31952296 PMCID: PMC7024268 DOI: 10.3390/molecules25020360] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 11/19/2022] Open
Abstract
The molecular chaperone Hsp90 is a ubiquitous ATPase-directed protein responsible for the activation and structural stabilization of a large clientele of proteins. As such, Hsp90 has emerged as a suitable candidate for the treatment of a diverse set of diseases, such as cancer and neurodegeneration. The inhibition of the chaperone through ATP-competitive inhibitors, however, was shown to lead to undesirable side effects. One strategy to alleviate this problem is the development of molecules that are able to disrupt specific protein–protein interactions, thus modulating the activity of Hsp90 only in the particular cellular pathway that needs to be targeted. Here, we exploit novel computational and theoretical approaches to design a set of peptides that are able to bind Hsp90 and compete for its interaction with the co-chaperone Cdc37, which is found to be responsible for the promotion of cancer cell proliferation. In spite of their capability to disrupt the Hsp90–Cdc37 interaction, no important cytotoxicity was observed in human cancer cells exposed to designed compounds. These findings imply the need for further optimization of the compounds, which may lead to new ways of interfering with the Hsp90 mechanisms that are important for tumour growth.
Collapse
|
8
|
Choi H, Kim HJ, Yang J, Chae S, Lee W, Chung S, Kim J, Choi H, Song H, Lee CK, Jun JH, Lee YJ, Lee K, Kim S, Sim H, Choi YI, Ryu KH, Park J, Lee D, Han S, Hwang D, Kyung J, Mook‐Jung I. Acetylation changes tau interactome to degrade tau in Alzheimer's disease animal and organoid models. Aging Cell 2020; 19:e13081. [PMID: 31763743 PMCID: PMC6974726 DOI: 10.1111/acel.13081] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/26/2019] [Accepted: 10/06/2019] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is an age‐related neurodegenerative disease. The most common pathological hallmarks are amyloid plaques and neurofibrillary tangles in the brain. In the brains of patients with AD, pathological tau is abnormally accumulated causing neuronal loss, synaptic dysfunction, and cognitive decline. We found a histone deacetylase 6 (HDAC6) inhibitor, CKD‐504, changed the tau interactome dramatically to degrade pathological tau not only in AD animal model (ADLPAPT) brains containing both amyloid plaques and neurofibrillary tangles but also in AD patient‐derived brain organoids. Acetylated tau recruited chaperone proteins such as Hsp40, Hsp70, and Hsp110, and this complex bound to novel tau E3 ligases including UBE2O and RNF14. This complex degraded pathological tau through proteasomal pathway. We also identified the responsible acetylation sites on tau. These dramatic tau‐interactome changes may result in tau degradation, leading to the recovery of synaptic pathology and cognitive decline in the ADLPAPT mice.
Collapse
Affiliation(s)
- Heesun Choi
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
| | - Haeng Jun Kim
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
| | - Jinhee Yang
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
- Department of Pharmacology CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Sehyun Chae
- Center for Plant Aging Research Institute for Basic Science DGIST Daegu Korea
- Korea Brain Bank Korea Brain Research Institute Daegu Korea
| | - Wonik Lee
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
| | - Sunwoo Chung
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
| | - Jisoo Kim
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
- Department of Pharmacology CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Hyunjung Choi
- Interdisciplinary Graduate Program in Genetic Engineering Seoul National University Seoul Korea
| | - Hyeseung Song
- Department of Medicinal Chemistry CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Chang Kon Lee
- Department of Medicinal Chemistry CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Jae Hyun Jun
- Department of Pharmacology CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Yong Jae Lee
- Department of Pharmacology CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Kyunghyeon Lee
- Department of Nonclinical Development CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Semi Kim
- Department of Nonclinical Development CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Hye‐ri Sim
- Department of Nonclinical Development CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Young Il Choi
- Department of Pharmacology CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Keun Ho Ryu
- Department of Nonclinical Development CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Jong‐Chan Park
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
| | - Dongjoon Lee
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
| | - Sun‐Ho Han
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
| | - Daehee Hwang
- Center for Plant Aging Research Institute for Basic Science DGIST Daegu Korea
- Department of New Biology DGIST Daegu Korea
| | - Jangbeen Kyung
- Department of Pharmacology CKD Research Institute CKD Pharmaceutical Company Seoul Korea
| | - Inhee Mook‐Jung
- Department of Biochemistry and Biomedical Sciences College of Medicine Seoul National University Seoul Korea
| |
Collapse
|
9
|
D'Annessa I, Raniolo S, Limongelli V, Di Marino D, Colombo G. Ligand Binding, Unbinding, and Allosteric Effects: Deciphering Small-Molecule Modulation of HSP90. J Chem Theory Comput 2019; 15:6368-6381. [PMID: 31538783 DOI: 10.1021/acs.jctc.9b00319] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The molecular chaperone HSP90 oversees the functional activation of a large number of client proteins. Because of its role in multiple pathways linked to cancer and neurodegeneration, drug discovery targeting HSP90 has been actively pursued. Yet, a number of inhibitors failed to meet expectations due to induced toxicity problems. In this context, allosteric perturbation has emerged as an alternative strategy for the pharmacological modulation of HSP90 functions. Specifically, novel allosteric stimulators showed the interesting capability of accelerating HSP90 closure dynamics and ATPase activities while inducing tumor cell death. Here, we gain atomistic insight into the mechanisms of allosteric ligand recognition and their consequences on the functional dynamics of HSP90, starting from the fully unbound state. We integrate advanced computational sampling methods based on FunnelMetadynamics, with the analysis of internal dynamics of the structural ensembles visited during the simulations. We observe several binding/unbinding events, and from these, we derive an accurate estimation of the absolute binding free energy. Importantly, we show that different binding poses induce different dynamics states. Our work for the first time explicitly correlates HSP90 responses to binding/unbinding of an allosteric ligand to the modulation of functionally oriented protein motions.
Collapse
Affiliation(s)
| | - Stefano Raniolo
- Università della Svizzera Italiana (USI) , Faculty of Biomedical Sciences, Institute of Computational Science - Center for Computational Medicine in Cardiology , via G. Buffi 13 , CH-Lugano , Switzerland
| | - Vittorio Limongelli
- Università della Svizzera Italiana (USI) , Faculty of Biomedical Sciences, Institute of Computational Science - Center for Computational Medicine in Cardiology , via G. Buffi 13 , CH-Lugano , Switzerland.,Department of Pharmacy , University of Naples ″Federico II″ , via D. Montesano 49 , I-80131 Naples , Italy
| | - Daniele Di Marino
- Università della Svizzera Italiana (USI) , Faculty of Biomedical Sciences, Institute of Computational Science - Center for Computational Medicine in Cardiology , via G. Buffi 13 , CH-Lugano , Switzerland.,Department of Life and Environmental Sciences - New York-Marche Structural Biology Center (NY-MaSBiC) , Polytechnic University of Marche , Via Brecce Bianche , 60131 Ancona , Italy
| | - Giorgio Colombo
- ICRM-CNR , Via Mario Bianco 9 , 20131 Milano , Italy.,Department of Chemistry , University of Pavia , V.le Taramelli 12 , 27100 Pavia , Italy
| |
Collapse
|
10
|
Cuadrado-Tejedor M, Pérez-González M, García-Muñoz C, Muruzabal D, García-Barroso C, Rabal O, Segura V, Sánchez-Arias JA, Oyarzabal J, Garcia-Osta A. Taking Advantage of the Selectivity of Histone Deacetylases and Phosphodiesterase Inhibitors to Design Better Therapeutic Strategies to Treat Alzheimer's Disease. Front Aging Neurosci 2019; 11:149. [PMID: 31281249 PMCID: PMC6597953 DOI: 10.3389/fnagi.2019.00149] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/06/2019] [Indexed: 12/19/2022] Open
Abstract
The discouraging results with therapies for Alzheimer’s disease (AD) in clinical trials, highlights the urgent need to adopt new approaches. Like other complex diseases, it is becoming clear that AD therapies should focus on the simultaneous modulation of several targets implicated in the disease. Recently, using reference compounds and the first-in class CM-414, we demonstrated that the simultaneous inhibition of histone deacetylases [class I histone deacetylases (HDACs) and HDAC6] and phosphodiesterase 5 (PDE5) has a synergistic therapeutic effect in AD models. To identify the best inhibitory balance of HDAC isoforms and PDEs that provides a safe and efficient therapy to combat AD, we tested the compound CM-695 in the Tg2576 mouse model of this disease. CM-695 selectively inhibits HDAC6 over class I HDAC isoforms, which largely overcomes the toxicity associated with HDAC class 1 inhibition. Furthermore, CM-695 inhibits PDE9, which is expressed strongly in the brain and has been proposed as a therapeutic target for AD. Chronic treatment of aged Tg2576 mice with CM-695 ameliorates memory impairment and diminishes brain Aβ, although its therapeutic effect was no longer apparent 4 weeks after the treatment was interrupted. An increase in the presence of 78-KDa glucose regulated protein (GRP78) and heat shock protein 70 (Hsp70) chaperones may underlie the therapeutic effect of CM-695. In summary, chronic treatment with CM-695 appears to reverse the AD phenotype in a safe and effective manner. Taking into account that AD is a multifactorial disorder, the multimodal action of these compounds and the different events they affect may open new avenues to combat AD.
Collapse
Affiliation(s)
- Mar Cuadrado-Tejedor
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Marta Pérez-González
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Cristina García-Muñoz
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Damián Muruzabal
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Carolina García-Barroso
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Obdulia Rabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Víctor Segura
- Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Bioinformatics Unit, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Juan A Sánchez-Arias
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Ana Garcia-Osta
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| |
Collapse
|
11
|
Martinelli AHS, Lopes FC, John EBO, Carlini CR, Ligabue-Braun R. Modulation of Disordered Proteins with a Focus on Neurodegenerative Diseases and Other Pathologies. Int J Mol Sci 2019; 20:ijms20061322. [PMID: 30875980 PMCID: PMC6471803 DOI: 10.3390/ijms20061322] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/03/2019] [Accepted: 02/12/2019] [Indexed: 12/15/2022] Open
Abstract
Intrinsically disordered proteins (IDPs) do not have rigid 3D structures, showing changes in their folding depending on the environment or ligands. Intrinsically disordered proteins are widely spread in eukaryotic genomes, and these proteins participate in many cell regulatory metabolism processes. Some IDPs, when aberrantly folded, can be the cause of some diseases such as Alzheimer′s, Parkinson′s, and prionic, among others. In these diseases, there are modifications in parts of the protein or in its entirety. A common conformational variation of these IDPs is misfolding and aggregation, forming, for instance, neurotoxic amyloid plaques. In this review, we discuss some IDPs that are involved in neurodegenerative diseases (such as beta amyloid, alpha synuclein, tau, and the “IDP-like” PrP), cancer (p53, c-Myc), and diabetes (amylin), focusing on the structural changes of these IDPs that are linked to such pathologies. We also present the IDP modulation mechanisms that can be explored in new strategies for drug design. Lastly, we show some candidate drugs that can be used in the future for the treatment of diseases caused by misfolded IDPs, considering that cancer therapy has more advanced research in comparison to other diseases, while also discussing recent and future developments in this area of research. Therefore, we aim to provide support to the study of IDPs and their modulation mechanisms as promising approaches to combat such severe diseases.
Collapse
Affiliation(s)
- Anne H S Martinelli
- Department of Molecular Biology and Biotechnology & Department of Biophysics, Biosciences Institute-IB, (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
| | - Fernanda C Lopes
- Center for Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
- Graduate Program in Cell and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
| | - Elisa B O John
- Center for Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
- Graduate Program in Cell and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
| | - Célia R Carlini
- Graduate Program in Cell and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
- Graduate Program in Medicine and Health Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre CEP 91410-000, RS, Brazil.
- Brain Institute-InsCer, Laboratory of Neurotoxins, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre CEP 90610-000, RS, Brazil.
| | - Rodrigo Ligabue-Braun
- Department of Pharmaceutical Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre CEP 90050-170, RS, Brazil.
| |
Collapse
|
12
|
Roe MS, Wahab B, Török Z, Horváth I, Vigh L, Prodromou C. Dihydropyridines Allosterically Modulate Hsp90 Providing a Novel Mechanism for Heat Shock Protein Co-induction and Neuroprotection. Front Mol Biosci 2018; 5:51. [PMID: 29930942 PMCID: PMC6000670 DOI: 10.3389/fmolb.2018.00051] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Chaperones play a pivotal role in protein homeostasis, but with age their ability to clear aggregated and damaged protein from cells declines. Tau pathology is a driver of a variety of neurodegenerative disease and in Alzheimer's disease (AD) it appears to be precipitated by the formation of amyloid-β (Aβ) aggregates. Aβ-peptide appears to trigger Tau hyperphosphorylation, formation of neurofibrillary tangles and neurotoxicity. Recently, dihydropyridine derivatives were shown to upregulate the heat shock response (HSR) and provide a neuroprotective effect in an APPxPS1 AD mouse model. The HSR response was only seen in diseased cells and consequently these compounds were defined as co-inducers since they upregulate chaperones and co-chaperones only when a pathological state is present. We show for compounds tested herein, that they target predominantly the C-terminal domain of Hsp90, but show some requirement for its middle-domain, and that binding stimulates the chaperones ATPase activity. We identify the site for LA1011 binding and confirm its identification by mutagenesis. We conclude, that binding compromises Hsp90's ability to chaperone, by modulating its ATPase activity, which consequently induces the HSR in diseased cells. Collectively, this represents the mechanism by which the normalization of neurofibrillary tangles, preservation of neurons, reduced tau pathology, reduced amyloid plaque, and increased dendritic spine density in the APPxPS1 Alzheimer's mouse model is initiated. Such dihydropyridine derivatives therefore represent potential pharmaceutical candidates for the therapy of neurodegenerative disease, such as AD.
Collapse
Affiliation(s)
- Mark S Roe
- Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom
| | - Ben Wahab
- Sussex Drug Discovery Centre, University of Sussex, Brighton, United Kingdom
| | - Zsolt Török
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences (HAS), Szeged, Hungary
| | - Ibolya Horváth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences (HAS), Szeged, Hungary
| | - László Vigh
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences (HAS), Szeged, Hungary
| | | |
Collapse
|
13
|
Kasza Á, Hunya Á, Frank Z, Fülöp F, Török Z, Balogh G, Sántha M, Bálind Á, Bernáth S, Blundell KLIM, Prodromou C, Horváth I, Zeiler HJ, Hooper PL, Vigh L, Penke B. Dihydropyridine Derivatives Modulate Heat Shock Responses and have a Neuroprotective Effect in a Transgenic Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2018; 53:557-71. [PMID: 27163800 PMCID: PMC4969717 DOI: 10.3233/jad-150860] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Heat shock proteins (Hsps) have chaperone activity and play a pivotal role in the homeostasis of proteins by preventing misfolding, by clearing aggregated and damaged proteins from cells, and by maintaining proteins in an active state. Alzheimer’s disease (AD) is thought to be caused by amyloid-β peptide that triggers tau hyperphosphorylation, which is neurotoxic. Although proteostasis capacity declines with age and facilitates the manifestation of neurodegenerative diseases such as AD, the upregulation of chaperones improves prognosis. Our research goal is to identify potent Hsp co-inducers that enhance protein homeostasis for the treatment of AD, especially 1,4-dihydropyridine derivatives optimized for their ability to modulate cellular stress responses. Based on favorable toxicological data and Hsp co-inducing activity, LA1011 was selected for the in vivo analysis of its neuroprotective effect in the APPxPS1 mouse model of AD. Here, we report that 6 months of LA1011 administration effectively improved the spatial learning and memory functions in wild type mice and eliminated neurodegeneration in double mutant mice. Furthermore, Hsp co-inducer therapy preserves the number of neurons, increases dendritic spine density, and reduces tau pathology and amyloid plaque formation in transgenic AD mice. In conclusion, the Hsp co-inducer LA1011 is neuroprotective and therefore is a potential pharmaceutical candidate for the therapy of neurodegenerative diseases, particularly AD.
Collapse
Affiliation(s)
- Ágnes Kasza
- Department of Medical Chemistry, University of Szeged, Hungary
| | - Ákos Hunya
- LipidArt Research and Development Ltd., Szeged, Hungary
| | - Zsuzsa Frank
- Department of Medical Chemistry, University of Szeged, Hungary
| | - Ferenc Fülöp
- Department of Pharmaceutical Chemistry, University of Szeged, Hungary
| | - Zsolt Török
- LipidArt Research and Development Ltd., Szeged, Hungary.,Biological Research Center of HAS, Institute of Biochemistry, Szeged, Hungary
| | - Gábor Balogh
- Biological Research Center of HAS, Institute of Biochemistry, Szeged, Hungary
| | - Miklós Sántha
- Biological Research Center of HAS, Institute of Biochemistry, Szeged, Hungary
| | - Árpád Bálind
- Biological Research Center of HAS, Institute of Biochemistry, Szeged, Hungary
| | | | | | | | - Ibolya Horváth
- Biological Research Center of HAS, Institute of Biochemistry, Szeged, Hungary
| | | | - Philip L Hooper
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Medical School, Anschutz Medical Campus, Aurora, CO, USA
| | - László Vigh
- Biological Research Center of HAS, Institute of Biochemistry, Szeged, Hungary
| | - Botond Penke
- Department of Medical Chemistry, University of Szeged, Hungary
| |
Collapse
|
14
|
Goode KM, Petrov DP, Vickman RE, Crist SA, Pascuzzi PE, Ratliff TL, Davisson VJ, Hazbun TR. Targeting the Hsp90 C-terminal domain to induce allosteric inhibition and selective client downregulation. Biochim Biophys Acta Gen Subj 2017; 1861:1992-2006. [PMID: 28495207 DOI: 10.1016/j.bbagen.2017.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 04/20/2017] [Accepted: 05/05/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inhibition of Hsp90 is desirable due to potential downregulation of oncogenic clients. Early generation inhibitors bind to the N-terminal domain (NTD) but C-terminal domain (CTD) inhibitors are a promising class because they do not induce a heat shock response. Here we present a new structural class of CTD binding molecules with a unique allosteric inhibition mechanism. METHODS A hit molecule, NSC145366, and structurally similar probes were assessed for inhibition of Hsp90 activities. A ligand-binding model was proposed indicating a novel Hsp90 CTD binding site. Client protein downregulation was also determined. RESULTS NSC145366 interacts with the Hsp90 CTD and has anti-proliferative activity in tumor cell lines (GI50=0.2-1.9μM). NSC145366 increases Hsp90 oligomerization resulting in allosteric inhibition of NTD ATPase activity (IC50=119μM) but does not compete with NTD or CTD-ATP binding. Treatment of LNCaP prostate tumor cells resulted in selective client protein downregulation including AR and BRCA1 but without a heat shock response. Analogs had similar potencies in ATPase and chaperone activity assays and variable effects on oligomerization. In silico modeling predicted a binding site at the CTD dimer interface distinct from the nucleotide-binding site. CONCLUSIONS A set of symmetrical scaffold molecules with bisphenol A cores induced allosteric inhibition of Hsp90. Experimental evidence and molecular modeling suggest that the binding site is independent of the CTD-ATP site and consistent with unique induction of allosteric effects. GENERAL SIGNIFICANCE Allosteric inhibition of Hsp90 via a mechanism used by the NSC145366-based probes is a promising avenue for selective oncogenic client downregulation.
Collapse
Affiliation(s)
- Kourtney M Goode
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Dino P Petrov
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Renee E Vickman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Scott A Crist
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Pete E Pascuzzi
- Purdue University Libraries Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Tim L Ratliff
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - V Jo Davisson
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Tony R Hazbun
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
15
|
Chebotareva N, Bobkova I, Shilov E. Heat shock proteins and kidney disease: perspectives of HSP therapy. Cell Stress Chaperones 2017; 22:319-343. [PMID: 28409327 PMCID: PMC5425374 DOI: 10.1007/s12192-017-0790-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/11/2017] [Accepted: 03/20/2017] [Indexed: 12/11/2022] Open
Abstract
Heat shock proteins (HSPs) mediate a diverse range of cellular functions, prominently including folding and regulatory processes of cellular repair. A major property of these remarkable proteins, dependent on intracellular or extracellular location, is their capacity for immunoregulation that optimizes immune activity while avoiding hyperactivated inflammation. In this review, recent investigations are described, which examine roles of HSPs in protection of kidney tissue from various traumatic influences and demonstrate their potential for clinical management of nephritic disease. The HSP70 class is particularly attractive in this respect due to its multiple protective effects. The review also summarizes current understanding of HSP bioactivity in the pathophysiology of various kidney diseases, including acute kidney injury, diabetic nephropathy, chronic glomerulonephritis, and lupus nephritis-along with other promising strategies for their remediation, such as DNA vaccination.
Collapse
Affiliation(s)
- Natalia Chebotareva
- I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya st., Moscow, Russia, 119992.
| | - Irina Bobkova
- I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya st., Moscow, Russia, 119992
| | - Evgeniy Shilov
- I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya st., Moscow, Russia, 119992
| |
Collapse
|