1
|
Mir IH, Shyam KT, Balakrishnan SS, Kumar MS, Ramesh T, Thirunavukkarasu C. Elucidation of escitalopram oxalate and related antidepressants as putative inhibitors of PTP4A3/PRL-3 protein in hepatocellular carcinoma: A multi-computational investigation. Comput Biol Chem 2024; 110:108039. [PMID: 38471352 DOI: 10.1016/j.compbiolchem.2024.108039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/12/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024]
Abstract
Hepatocellular carcinoma (HCC) persists to be one of the most devastating and deadliest malignancies globally. Recent research into the molecular signaling networks entailed in many malignancies has given some prominent insights that can be leveraged to create molecular therapeutics for combating HCC. Therefore, in the current communication, an in-silico drug repurposing approach has been employed to target the function of PTP4A3/PRL-3 protein in HCC using antidepressants: Fluoxetine hydrochloride, Citalopram, Amitriptyline, Imipramine, and Escitalopram oxalate as the desired ligands. The density function theory (DFT) and chemical absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters for the chosen ligands were evaluated to comprehend the pharmacokinetics, drug-likeness properties, and bioreactivity of the ligands. The precise interaction mechanism was explored using computational methods such as molecular docking and molecular dynamics (MD) simulation studies to assess the inhibitory effect and the stability of the interactions against the protein of interest. Escitalopram oxalate exhibited a comparatively significant docking score (-7.4 kcal/mol) compared to the control JMS-053 (-6.8 kcal/mol) against the PRL-3 protein. The 2D interaction plots exhibited an array of hydrophobic and hydrogen bond interactions. The findings of the ADMET forecast confirmed that it adheres to Lipinski's rule of five with no violations, and DFT analysis revealed a HOMO-LUMO energy gap of -0.26778 ev, demonstrating better reactivity than the control molecule. The docked complexes were subjected to MD studies (100 ns) showing stable interactions. Considering all the findings, it can be concluded that Escitalopram oxalate and related therapeutics can act as potential pharmacological candidates for targeting the activity of PTP4A3/PRL-3 in HCC.
Collapse
Affiliation(s)
- Ishfaq Hassan Mir
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605 014, India
| | - Kankipati Teja Shyam
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605 014, India
| | | | | | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | |
Collapse
|
2
|
Bhavana, Kohal R, Kumari P, Das Gupta G, Kumar Verma S. Druggable targets of protein tyrosine phosphatase Family, viz. PTP1B, SHP2, Cdc25, and LMW-PTP: Current scenario on medicinal Attributes, and SAR insights. Bioorg Chem 2024; 144:107121. [PMID: 38237392 DOI: 10.1016/j.bioorg.2024.107121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/02/2024] [Accepted: 01/10/2024] [Indexed: 02/17/2024]
Abstract
Protein tyrosine phosphatases (PTPs) are the class of dephosphorylation enzymes that catalyze the removal of phosphate groups from tyrosine residues on proteins responsible for various cellular processes. Any disbalance in signal pathways mediated by PTPs leads to various disease conditions like diabetes, obesity, cancers, and autoimmune disorders. Amongst the PTP superfamily, PTP1B, SHP2, Cdc25, and LMW-PTP have been prioritized as druggable targets for developing medicinal agents. PTP1B is an intracellular PTP enzyme that downregulates insulin and leptin signaling pathways and is involved in insulin resistance and glucose homeostasis. SHP2 is involved in the RAS-MAPK pathway and T cell immunity. Cdk-cyclin complex activation occurs by Cdc25-PTPs involved in cell cycle regulation. LMW-PTPs are involved in PDGF/PDGFR, Eph/ephrin, and insulin signaling pathways, resulting in certain diseases like diabetes mellitus, obesity, and cancer. The signaling cascades of PTP1B, SHP2, Cdc25, and LMW-PTPs have been described to rationalize their medicinal importance in the pathophysiology of diabetes, obesity, and cancer. Their binding sites have been explored to overcome the hurdles in discovering target selective molecules with optimum potency. Recent developments in the synthetic molecules bearing heterocyclic moieties against these targets have been explored to gain insight into structural features. The elaborated SAR investigation revealed the effect of substituents on the potency and target selectivity, which can be implicated in the further discovery of newer medicinal agents targeting the druggable members of the PTP superfamily.
Collapse
Affiliation(s)
- Bhavana
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, (Punjab), India
| | - Rupali Kohal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, (Punjab), India
| | - Preety Kumari
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, (Punjab), India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga 142 001, (Punjab), India
| | - Sant Kumar Verma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, (Punjab), India.
| |
Collapse
|
3
|
Xie WJ, Liu M, Zhang X, Zhang YG, Jian ZH, Xiong XX. Astaxanthin suppresses LPS-induced myocardial apoptosis by regulating PTP1B/JNK pathway in vitro. Int Immunopharmacol 2024; 127:111395. [PMID: 38141411 DOI: 10.1016/j.intimp.2023.111395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 12/25/2023]
Abstract
PURPOSE Myocardial injury induced by sepsis can increase the patient's mortality, which is an important complication of sepsis. Myocardial apoptosis plays a key role in septic myocardial injury. Here we explored the potential mechanism of astaxanthin (ATX) inhibiting myocardial apoptosis induced by lipopolysaccharide (LPS) in vitro. METHODS The H9C2 cell experiment was conducted in three parts. In the first part, we set up three groups: control group, LPS group (10 µg/ml), a model of septic myocardial injury, and LPS + ATX (5, 10, 30 µM); In the second part, we set up four groups: control group, LPS group, LPS + PTP1B-IN-1, a protein tyrosine phosphatase 1B (PTP1B) inhibitor, and LPS + PTP1B-IN-1 + ATX; In the third part, we set up four groups: control group, LPS group, LPS + Anisomycin, a c-Jun N-terminal kinase (JNK) activator, and LPS + Anisomycin + ATX. We assessed H9C2 cell viability using the Cell Counting Kit-8 (CCK-8) assay. We observed cell apoptosis using flow cytometry analysis. We tested the mitochondrial membrane potential (ΔΨm) using JC-1 staining. To identify the molecular targets of ATX, Astaxanthin targets were predicted through the SwissTargetPrediction database. We verified the binding affinity of ATX and its targets using microscale thermophoresis (MST). We investigated the p-JNK expression using immunofluorescence staining. Finally, Western blot was used to evaluate PTP1B, JNK, p-JNK and the mitochondrial apoptosis-associated protein expression. RESULTS LPS inhibited H9C2 cell viability in a time-dependent manner and ATX treatment enhances H9C2 cell viability in a concentration dependent manner after LPS administration. ATX inhibited the LPS-induced apoptosis and loss of mitochondrial membrane potential in H9C2 cells. As predicted by the SwissTargetPrediction database, PTP1B was a potential target of ATX, and the interaction between ATX and PTP1B was further verified by MST. ATX attenuated the LPS-induced protein expression of PTP1B and p-JNK, regardless of PTP1B inhibition. Both immunofluorescence staining and Western blotting showed that ATX suppressed the LPS-induced p-JNK expression in H9C2 cells, regardless of Anisomycin administration. In addition, by adding Anisomycin to overexpress JNK, ATX inhibited the LPS-induced apoptosis, loss of mitochondrial membrane potential and upregulation of mitochondrial apoptosis-associated proteins in H9C2 cells via JNK signaling. CONCLUSION ATX inhibited LPS-induced mitochondrial apoptosis of H9C2 cells by PTP1B/JNK pathway and PTP1B was the target of ATX.
Collapse
Affiliation(s)
- Wen-Jie Xie
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Miao Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xu Zhang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yong-Gang Zhang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhi-Hong Jian
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Xiao-Xing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
4
|
Qasem B, Dąbrowska A, Króliczewski J, Łyczko J, Marycz K. Trodusquemine (MSI-1436) Restores Metabolic Flexibility and Mitochondrial Dynamics in Insulin-Resistant Equine Hepatic Progenitor Cells (HPCs). Cells 2024; 13:152. [PMID: 38247843 PMCID: PMC10814577 DOI: 10.3390/cells13020152] [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: 11/18/2023] [Revised: 12/31/2023] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Equine metabolic syndrome (EMS) is a significant global health concern in veterinary medicine. There is increasing interest in utilizing molecular agents to modulate hepatocyte function for potential clinical applications. Recent studies have shown promising results in inhibiting protein tyrosine phosphatase (PTP1B) to maintain cell function in various models. In this study, we investigated the effects of the inhibitor Trodusquemine (MSI-1436) on equine hepatic progenitor cells (HPCs) under lipotoxic conditions. We examined proliferative activity, glucose uptake, and mitochondrial morphogenesis. Our study found that MSI-1436 promotes HPC entry into the cell cycle and protects them from palmitate-induced apoptosis by regulating mitochondrial dynamics and biogenesis. MSI-1436 also increases glucose uptake and protects HPCs from palmitate-induced stress by reorganizing the cells' morphological architecture. Furthermore, our findings suggest that MSI-1436 enhances 2-NBDG uptake by increasing the expression of SIRT1, which is associated with liver insulin sensitivity. It also promotes mitochondrial dynamics by modulating mitochondria quantity and morphotype as well as increasing the expression of PINK1, MFN1, and MFN2. Our study provides evidence that MSI-1436 has a positive impact on equine hepatic progenitor cells, indicating its potential therapeutic value in treating EMS and insulin dysregulation.
Collapse
Affiliation(s)
- Badr Qasem
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; (B.Q.); (A.D.); (J.K.)
| | - Agnieszka Dąbrowska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; (B.Q.); (A.D.); (J.K.)
| | - Jarosław Króliczewski
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; (B.Q.); (A.D.); (J.K.)
| | - Jacek Łyczko
- Department of Food Chemistry and Biocatalysis, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland;
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; (B.Q.); (A.D.); (J.K.)
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA 95516, USA
| |
Collapse
|
5
|
Patel K, Bora V, Patel B. Sodium orthovanadate exhibits anti-angiogenic, antiapoptotic and blood glucose-lowering effect on colon cancer associated with diabetes. Cancer Chemother Pharmacol 2024; 93:55-70. [PMID: 37755518 DOI: 10.1007/s00280-023-04596-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND The presence of type 2 diabetes mellitus increases the risk of developing the colon cancer. The main objective of this study was to determine the role of sodium orthovanadate (SOV) in colon cancer associated with diabetes mellitus by targeting the competitive inhibition of PTP1B. METHODS For in vivo study, high fat diet with low dose streptozotocin model was used for inducing the diabetes mellitus. Colon cancer was induced by injecting 1,2-dimethylhydrazine (25 mg/kg, sc) twice a week. TNM staging and immunohistochemistry (IHC) was carried out for colon cancer tissues. In vitro studies like MTT assay, clonogenic assay, rhodamine-123 dye assay and annexin V-FITC assay using flow cytometry were performed on HCT-116 cell line. CAM assay was performed to examine the anti-angiogenic effect of the drug. RESULTS Sodium orthovanadate reduces the blood glucose level and tumor parameters in the animals. In vitro studies revealed that SOV decreased cell proliferation dose dependently. In addition, SOV induced apoptosis as depicted from rhodamine-123 dye assay and annexin V-FITC assay using flow cytometry as well as p53 IHC staining. SOV showed reduced angiogenesis effect on eggs which was depicted from CAM assay and also from CD34 and E-cadherin IHC staining. CONCLUSIONS Our data suggest that SOV exhibits protective role in colon cancer associated with diabetes mellitus. SOV exhibits anti-proliferative, anti-angiogenic and apoptotic inducing effects hence can be considered for therapeutic switching in diabetic colon cancer.
Collapse
Affiliation(s)
- Kruti Patel
- Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Vivek Bora
- Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Bhoomika Patel
- National Forensic Sciences University, Sector 9, Gandhinagar, 382007, Gujarat, India.
| |
Collapse
|
6
|
Yuan BY, Zhuang Y, Wu ZF, Zhao XM, Zhang L, Chen GW, Zeng ZC. miR-146a-5p Alleviates Radiation-Induced Liver Fibrosis by Regulating PTPRA-SRC Signaling in Mice. Radiat Res 2023; 200:531-537. [PMID: 38014555 DOI: 10.1667/rade-22-00017.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 10/05/2023] [Indexed: 11/29/2023]
Abstract
Patients with hepatobiliary tumors who accept radiotherapy are at risk for radiation-induced liver fibrosis. MicroRNAs (miRNAs) have been implicated in the pathogenesis of radiation-induced liver damage and possess potential as novel biomarkers and therapeutic targets. However, the role of miR-146a-5p in radiation-induced liver fibrosis is less well understood. The current study was designed to evaluate the role of miR-146a-5p in radiation-induced liver fibrosis in mice and to investigate the possible mechanisms involved in miR-146a-5p-mediated effects. The experiments were performed on Institute of Cancer Research (ICR) mice which received fractionated radiation (30 Gy in 5 fractions) to the liver. The results show radiation could induce histopathological changes, liver dysfunction and fibrosis accompanied with decreased miR-146a-5p expression. miR-146a-5p agomir treatment resulted in recovery of liver function and reduced the amount of alpha-smooth muscle actin (α-SMA), collagen 1, protein tyrosine phosphatase receptor type A (PTPRA) and phosphorylated SRC in the livers of irradiated mice. Therefore, our study reveals that miR-146a-5p inhibits the progression of hepatic fibrosis after radiation treatment. And the beneficial role of miR-146a-5p may be relevant to PTPRA-SRC signaling pathway.
Collapse
Affiliation(s)
- Bao-Ying Yuan
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Zhuang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhi-Feng Wu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Mei Zhao
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gen-Wen Chen
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Yang Q, Zou Y, Wei X, Ye P, Wu Y, Ai H, Zhang Z, Tan J, Zhou J, Yang Y, Dai Q, Dou C, Luo F. PTP1B knockdown alleviates BMSCs senescence via activating AMPK-mediated mitophagy and promotes osteogenesis in senile osteoporosis. Biochim Biophys Acta Mol Basis Dis 2023:166795. [PMID: 37385514 DOI: 10.1016/j.bbadis.2023.166795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
The senescence of bone marrow mesenchymal stem cells (BMSCs) is the basis of senile osteoporosis (SOP). Targeting BMSCs senescence is of paramount importance for developing anti-osteoporotic strategy. In this study, we found that protein tyrosine phosphatase 1B (PTP1B), an enzyme responsible for tyrosine dephosphorylation, was significantly upregulated in BMSCs and femurs with advancing chronological age. Therefore, the potential role of PTP1B in BMSCs senescence and senile osteoporosis was studied. Firstly, significantly upregulated PTP1B expression along with impaired osteogenic differentiation capacity was observed in D-galactose (D-gal)-induced BMSCs and naturally-aged BMSCs. Furthermore, PTP1B silencing could effectively alleviate senescence, improve mitochondrial dysfunction, and restore osteogenic differentiation in aged BMSCs, which was attributable to enhanced mitophagy mediated by PKM2/AMPK pathway. In addition, hydroxychloroquine (HCQ), an autophagy inhibitor, significantly reversed the protective effects from PTP1B knockdown. In SOP animal model, transplantation of LVsh-PTP1B-transfected D-gal-induced BMSCs harvested double protective effects, including increased bone formation and reduced osteoclastogenesis. Similarly, HCQ treatment remarkably suppressed osteogenesis of LVsh-PTP1B-transfected D-gal-induced BMSCs in vivo. Taken together, our data demonstrated that PTP1B silencing protects against BMSCs senescence and mitigates SOP via activating AMPK-mediated mitophagy. Targeting PTP1B may represent a promising interventional strategy to attenuate SOP.
Collapse
Affiliation(s)
- QianKun Yang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuChi Zou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - XiaoYu Wei
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peng Ye
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuTong Wu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - HongBo Ai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhao Zhang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Orthopedics Department, The General Hospital of Western Theater Command PLA, Chengdu 610083, Sichuan Province, China
| | - JiuLin Tan
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiangling Zhou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuSheng Yang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - QiJie Dai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ce Dou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Fei Luo
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
8
|
Zhang M, Liu C, Zhao L, Zhang X, Su Y. The Emerging Role of Protein Phosphatase in Regeneration. Life (Basel) 2023; 13:life13051216. [PMID: 37240861 DOI: 10.3390/life13051216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Maintaining normal cellular behavior is essential for the survival of organisms. One of the main mechanisms to control cellular behavior is protein phosphorylation. The process of protein phosphorylation is reversible under the regulation of protein kinases and protein phosphatases. The importance of kinases in numerous cellular processes has been well recognized. In recent years, protein phosphatases have also been demonstrated to function actively and specifically in various cellular processes and thus have gained more and more attention from researchers. In the animal kingdom, regeneration frequently occurs to replace or repair damaged or missing tissues. Emerging evidence has revealed that protein phosphatases are crucial for organ regeneration. In this review, after providing a brief overview of the classification of protein phosphatases and their functions in several representative developmental processes, we highlight the critical roles that protein phosphatases play in organ regeneration by summarizing the most recent research on the function and underlying mechanism of protein phosphatase in the regeneration of the liver, bone, neuron, and heart in vertebrates.
Collapse
Affiliation(s)
- Meiling Zhang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Chenglin Liu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Long Zhao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Xuejiao Zhang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
9
|
Zhang ZH, Zhou XM, Zhang X. Role of Protein Tyrosine Phosphatase 1B Inhibitor in Early Brain Injury of Subarachnoid Hemorrhage in Mice. Brain Sci 2023; 13:brainsci13050816. [PMID: 37239288 DOI: 10.3390/brainsci13050816] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Clinically, early brain injury (EBI), which refers to the acute injuries to the whole brain in the phase of the first 72 h following subarachnoid hemorrhage (SAH), is intensely investigated to improve neurological and psychological function. Additionally, it will be meaningful to explore new therapeutic approaches for EBI treatment to improve the prognosis of patients with SAH. To investigate the underlying neuroprotection mechanism in vitro, the Protein tyrosine phosphatase 1B inhibitor (PTP1B-IN-1) was put in primary neurons induced by OxyHb to observe neuroapoptosis, neuroinflammation, and ER stress. Then, one hundred forty male mice were subjected to Experiment two and Experiment three. The mice in the SAH24h + PTP1B-IN-1 group were given an intraperitoneal injection of 5 mg/kg PTP1B-IN-1 30 min before anesthesia. SAH grade, neurological score, brain water content, Western blot, PCR, and Transmission Electron Microscopy (TEM) were performed to observe the underlying neuroprotection mechanism in vivo. Overall, this study suggests that PTP1B-IN-1 could ameliorate neuroapoptosis, neuroinflammation, and ER stress in vitro and in vivo by regulating the IRS-2/AKT signaling pathway, suggesting that PTP1B-IN-1 may be a candidate drug for the treatment of early brain injury after SAH.
Collapse
Affiliation(s)
- Zhong-Hua Zhang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
- Department of Anesthesiology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
| | - Xiao-Ming Zhou
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
| |
Collapse
|
10
|
Li N, Li X, Deng M, Zhu F, Wang Z, Sheng R, Wu W, Guo R. Isosteviol derivatives as protein tyrosine Phosphatase-1B inhibitors: Synthesis, biological evaluation and molecular docking. Bioorg Med Chem 2023; 83:117240. [PMID: 36963270 DOI: 10.1016/j.bmc.2023.117240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/02/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023]
Abstract
Protein tyrosine phosphatase (PTP1B) antagonizes insulin signaling and acts as a potential therapeutic target for insulin resistance associated with obesity and type II diabetes. In this work, a series of isosteviol derivatives 1-28 was synthesized and the inhibitory activity on PTP1B was evaluated by double antibody sandwich ELISA (DAS-ELISA) in vitro. Most isosteviol derivatives showed moderate PTP1B inhibitory activities. Among them, derivatives 10, 13, 24, 27 showed remarkable bioactivities with IC50 values ranging from 0.24 to 0.40 µM. Particularly, derivative 24 exhibited the best inhibitory activity against PTP1B (IC50 = 0.24 µM) in vitro; moreover, it showed 7-fold selectivity to PTP1B over T-cell protein tyrosine phosphatase (TCPTP) and 14-fold selectivity to PTP1B over cell division cycle 25 homolog B (CDC25B). Molecular docking studies demonstrated the hydrogen bond interaction between 24 and LYS-116 residue in PTP1B might be essential for the inhibitory activity. The results suggested that derivative 24 has great potential to be employed as drug candidate for the treatment of obesity and type II diabetes.
Collapse
Affiliation(s)
- Na Li
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Xinyu Li
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Meidi Deng
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Feifei Zhu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Zian Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Ruilong Sheng
- CQM-Centro de Química da Madeira, Campus da Penteada, Universidade da Madeira, 9000-390 Funchal, Portugal
| | - Wenhui Wu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Ruihua Guo
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai 201306, China; Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai 201306, China.
| |
Collapse
|
11
|
Behl T, Gupta A, Sehgal A, Albarrati A, Albratty M, Meraya AM, Najmi A, Bhatia S, Bungau S. Exploring protein tyrosine phosphatases (PTP) and PTP-1B inhibitors in management of diabetes mellitus. Biomed Pharmacother 2022; 153:113405. [DOI: 10.1016/j.biopha.2022.113405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/02/2022] Open
|
12
|
Lee WY, Lee CY, Lee JS, Kim CE. Identifying Candidate Flavonoids for Non-Alcoholic Fatty Liver Disease by Network-Based Strategy. Front Pharmacol 2022; 13:892559. [PMID: 35721123 PMCID: PMC9204489 DOI: 10.3389/fphar.2022.892559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/22/2022] [Indexed: 11/16/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common type of chronic liver disease and lacks guaranteed pharmacological therapeutic options. In this study, we applied a network-based framework for comprehensively identifying candidate flavonoids for the prevention and/or treatment of NAFLD. Flavonoid-target interaction information was obtained from combining experimentally validated data and results obtained using a recently developed machine-learning model, AI-DTI. Flavonoids were then prioritized by calculating the network proximity between flavonoid targets and NAFLD-associated proteins. The preventive effects of the candidate flavonoids were evaluated using FFA-induced hepatic steatosis in HepG2 and AML12 cells. We reconstructed the flavonoid-target network and found that the number of re-covered compound-target interactions was significantly higher than the chance level. Proximity scores have successfully rediscovered flavonoids and their potential mechanisms that are reported to have therapeutic effects on NAFLD. Finally, we revealed that discovered candidates, particularly glycitin, significantly attenuated lipid accumulation and moderately inhibited intracellular reactive oxygen species production. We further confirmed the affinity of glycitin with the predicted target using molecular docking and found that glycitin targets are closely related to several proteins involved in lipid metabolism, inflammatory responses, and oxidative stress. The predicted network-level effects were validated at the levels of mRNA. In summary, our study offers and validates network-based methods for the identification of candidate flavonoids for NAFLD.
Collapse
Affiliation(s)
- Won-Yung Lee
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam, South Korea
- Department of Herbal Formula, College of Korean Medicine, Dongguk University, Goyang-si, South Korea
| | - Choong-Yeol Lee
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam, South Korea
| | - Jin-Seok Lee
- Institute of Bioscience and Integrative Medicine, Daejeon Oriental Hospital of Daejeon University, Daejeon, South Korea
| | - Chang-Eop Kim
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam, South Korea
| |
Collapse
|
13
|
Busquets O, Espinosa-Jiménez T, Ettcheto M, Olloquequi J, Bulló M, Carro E, Cantero JL, Casadesús G, Folch J, Verdaguer E, Auladell C, Camins A. JNK1 and JNK3: divergent functions in hippocampal metabolic-cognitive function. Mol Med 2022; 28:48. [PMID: 35508978 PMCID: PMC9066854 DOI: 10.1186/s10020-022-00471-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/08/2022] [Indexed: 11/21/2022] Open
Abstract
Background and aim The appearance of alterations in normal metabolic activity has been increasingly considered a risk factor for the development of sporadic and late-onset neurodegenerative diseases. In this report, we induced chronic metabolic stress by feeding of a high-fat diet (HFD) in order to study its consequences in cognition. We also studied the effects of a loss of function of isoforms 1 and 3 of the c-Jun N-terminal Kinases (JNK), stress and cell death response elements. Methods Animals were fed either with conventional chow or with HFD, from their weaning until their sacrifice at 9 months. Before sacrifice, body weight, intraperitoneal glucose and insulin tolerance test (IP-GTT and IP‑ITT) were performed to evaluate peripheral biometrics. Additionally, cognitive behavioral tests and analysis of spine density were performed to assess cognitive function. Molecular studies were carried out to confirm the effects of metabolic stressors in the hippocampus relative to cognitive loss. Results Our studies demonstrated that HFD in Jnk3−/− lead to synergetic responses. Loss of function of JNK3 led to increased body weight, especially when exposed to an HFD and they had significantly decreased response to insulin. These mice also showed increased stress in the endoplasmic reticulum and diminished cognitive capacity. However, loss of function of JNK1 promoted normal or heightened energetic metabolism and preserved cognitive function even when chronically metabolically stressed. Conclusions Downregulation of JNK3 does not seem to be a suitable target for the modulation of energetic-cognitive dysregulations while loss of function of JNK1 seems to promote a good metabolic-cognitive profile, just like resistance to the negative effects of chronic feeding with HFD. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00471-y.
Collapse
Affiliation(s)
- Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, 08028, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Medicine and Health Sciences Faculty, University Rovira i Virgili, 43201, Reus, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain.,Institut de Neurociències, University of Barcelona, 08035, Barcelona, Spain.,Dominick P. Purpura Department of Neurosciences, Albert Einstein College of Medicine, New York City, 10461, USA
| | - Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, 08028, Barcelona, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain.,Institut de Neurociències, University of Barcelona, 08035, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, 08028, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Medicine and Health Sciences Faculty, University Rovira i Virgili, 43201, Reus, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain.,Institut de Neurociències, University of Barcelona, 08035, Barcelona, Spain
| | - Jordi Olloquequi
- Laboratory of Cellular and Molecular Pathology, Facultad de Ciencias de La Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Mònica Bulló
- Department of Biochemistry and Biotechnology, Medicine and Health Sciences Faculty, University Rovira i Virgili, 43201, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Sant Joan de Reus, 43204, Reus, Spain.,CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Eva Carro
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - José Luis Cantero
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain.,Laboratory of Functional Neuroscience, Pablo de Olavide University, 41013, Seville, Spain
| | - Gemma Casadesús
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Jaume Folch
- Department of Biochemistry and Biotechnology, Medicine and Health Sciences Faculty, University Rovira i Virgili, 43201, Reus, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Ester Verdaguer
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain.,Institut de Neurociències, University of Barcelona, 08035, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, University of Barcelona, 08028, Barcelona, Spain
| | - Carme Auladell
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain.,Institut de Neurociències, University of Barcelona, 08035, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, University of Barcelona, 08028, Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, 08028, Barcelona, Spain. .,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain. .,Institut de Neurociències, University of Barcelona, 08035, Barcelona, Spain.
| |
Collapse
|
14
|
Kim HJ, Ryu KY, Kim YG, Kim MO, Lee JH, Song MK, Youn YJ, Pokhrel NK, Kim SH, Kim JY, Jung HJ, Kim WS, Hong CW, Kim HH, Lee Y. Myeloid-Specific PTP1B Deficiency Attenuates Inflammation-Induced and Ovariectomy-Induced Bone Loss in Mice by Inhibiting Osteoclastogenesis. J Bone Miner Res 2022; 37:505-514. [PMID: 34812548 DOI: 10.1002/jbmr.4478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/09/2021] [Accepted: 11/17/2021] [Indexed: 11/11/2022]
Abstract
The differentiation and activity of bone-resorbing osteoclasts are tightly regulated to maintain the homeostasis of healthy bones. In this study, the role of protein tyrosine phosphatase 1B (PTP1B) during osteoclastogenesis was studied in myeloid-specific Ptpn1-deficient (conditional knockout [cKO]) mice. The mRNA and protein expression of PTP1B increased during the formation of mature osteoclasts from mouse bone macrophages on stimulation with macrophage-colony stimulating factor (M-CSF) and receptor activator of nuclear factor κB ligand (RANKL). The Ptpn1 cKO mice exhibited increased femoral trabecular bone volume with a decreased number and activity of osteoclasts compared with control mice. The in vitro culture of osteoclast precursors corroborated the inhibition of osteoclastogenesis in cKO cells compared with control, with concomitantly decreased RANKL-dependent proliferation, lower osteoclast marker gene expression, reduced nuclear expression of nuclear factor of activated T cells cytoplasmic 1 (NFATc1), diminished intracellular Ca2+ oscillations, and increased phosphorylation of proto-oncogene tyrosine-protein kinase Src on inhibitory tyrosine residue. In a ligature-induced periodontitis model, Ptpn1 cKO mice exhibited attenuated osteoclastogenesis and alveolar bone loss following the induction of inflammation. The Ptpn1-deficient mice were similarly protected from ovariectomy-induced bone loss compared with control mice. These results provide a novel regulatory role of PTP1B in osteoclastogenesis and suggest a potential as a therapeutic target for bone-lytic diseases. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Hyo Jeong Kim
- Department of Biochemistry and Institute for Hard Tissue and Bone Regeneration, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Ka-Young Ryu
- Department of Biochemistry and Institute for Hard Tissue and Bone Regeneration, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Yong-Gun Kim
- Department of Periodontology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Myoung Ok Kim
- Department of Animal Biotechnology, College of Ecology and Environmental Science, Kyungpook National University, Sangju, South Korea
| | - Ji Hye Lee
- Department of Oral Pathology, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Min-Kyoung Song
- Department of Cell and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Young-Jin Youn
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Nitin Kumar Pokhrel
- Department of Biochemistry and Institute for Hard Tissue and Bone Regeneration, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Sung-Hyun Kim
- Department of Bio-medical Analysis, Korea Polytechnic College, Chungnam, South Korea
| | - Jae-Young Kim
- Department of Biochemistry and Institute for Hard Tissue and Bone Regeneration, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Hye-Jin Jung
- Department of Biochemistry and Institute for Hard Tissue and Bone Regeneration, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Woo-Shin Kim
- Department of Cell and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Chang-Won Hong
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Youngkyun Lee
- Department of Biochemistry and Institute for Hard Tissue and Bone Regeneration, School of Dentistry, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
15
|
Fernández C, Torrealba N, Altamirano F, Garrido-Moreno V, Vásquez-Trincado C, Flores-Vergara R, López-Crisosto C, Ocaranza MP, Chiong M, Pedrozo Z, Lavandero S. Polycystin-1 is required for insulin-like growth factor 1-induced cardiomyocyte hypertrophy. PLoS One 2021; 16:e0255452. [PMID: 34407099 PMCID: PMC8372926 DOI: 10.1371/journal.pone.0255452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/18/2021] [Indexed: 11/19/2022] Open
Abstract
Cardiac hypertrophy is the result of responses to various physiological or pathological stimuli. Recently, we showed that polycystin-1 participates in cardiomyocyte hypertrophy elicited by pressure overload and mechanical stress. Interestingly, polycystin-1 knockdown does not affect phenylephrine-induced cardiomyocyte hypertrophy, suggesting that the effects of polycystin-1 are stimulus-dependent. In this study, we aimed to identify the role of polycystin-1 in insulin-like growth factor-1 (IGF-1) signaling in cardiomyocytes. Polycystin-1 knockdown completely blunted IGF-1-induced cardiomyocyte hypertrophy. We then investigated the molecular mechanism underlying this result. We found that polycystin-1 silencing impaired the activation of the IGF-1 receptor, Akt, and ERK1/2 elicited by IGF-1. Remarkably, IGF-1-induced IGF-1 receptor, Akt, and ERK1/2 phosphorylations were restored when protein tyrosine phosphatase 1B was inhibited, suggesting that polycystin-1 knockdown deregulates this phosphatase in cardiomyocytes. Moreover, protein tyrosine phosphatase 1B inhibition also restored IGF-1-dependent cardiomyocyte hypertrophy in polycystin-1-deficient cells. Our findings provide the first evidence that polycystin-1 regulates IGF-1-induced cardiomyocyte hypertrophy through a mechanism involving protein tyrosine phosphatase 1B.
Collapse
Affiliation(s)
- Carolina Fernández
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Natalia Torrealba
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
- Laboratory of Tumour Resistance, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Francisco Altamirano
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Cardiovascular Sciences, DeBakey Heart & Vascular Center Houston Methodist Research Institute, Houston, Texas, United States of America
- Department of Cardiothoracic Surgery, Weill Cornell Medical College, Cornell University, Ithaca, New York, United States of America
| | - Valeria Garrido-Moreno
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - César Vásquez-Trincado
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Raúl Flores-Vergara
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
- Facultad de Medicina, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago de Chile, Chile
| | - Camila López-Crisosto
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
- Faculty of Medicine, Division of Cardiovascular Diseases, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - María Paz Ocaranza
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
- Faculty of Medicine, Division of Cardiovascular Diseases, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
- Center for New Drugs for Hypertension (CENDHY), Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Mario Chiong
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Zully Pedrozo
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
- Facultad de Medicina, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago de Chile, Chile
| | - Sergio Lavandero
- Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Santiago de Chile, Chile
| |
Collapse
|
16
|
Bourebaba N, Marycz K. Hepatic stellate cells role in the course of metabolic disorders development - A molecular overview. Pharmacol Res 2021; 170:105739. [PMID: 34171492 DOI: 10.1016/j.phrs.2021.105739] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/07/2021] [Accepted: 06/19/2021] [Indexed: 02/08/2023]
Abstract
Fibrosis is characterized by an abnormal accumulation of extracellular matrix (ECM) constituents in the liver parenchyma that lead to hepatic cirrhosis. After liver injury, the hepatic stellate cells (HSCs) undergo a response called "activation", transforming the cells into proliferative, fibrogenic and contractile myofibroblasts, representing the main collagen-producing cells in the injured tissue. Activated HSCs are considered as pro-inflammatory cells producing cytokines and several hepatomatogens; they are additionally involved in the recruitment of Kupffer cells, circulating monocytes and macrophages through the production of chemokines. Moreover, HSC have been proposed as being involved in the development of insulin resistance mainly mediated by their inflammatory properties, which undeniably links their activation to the development of diabetes and Non-alcoholic fatty liver disease. In addition, when the liver is injured, a complex interaction between hepatocytes and HSCs occurs, inducing mitochondrial dysfunction, which contributes to the accumulation of fats in hepatocytes that trigger to liver lipotoxicity. These mechanisms underlying the activation of HSC suggest their major role in the development of metabolic disorders. It turns out that several molecules including MicroRNAs and proteins have the ability to inhibit the activation and the proliferation of HSCs, which makes them interesting therapeutic targets for the subsequent management of metabolic conditions. This review focuses on the mechanisms and molecular pathways underlying the initiation and onset of metabolic disorders following HSCs activation, as well as on molecular therapeutic targets, which could limit their fibrogenic transdifferentiation and therefore improve the liver condition in the course of metabolic imbalance.
Collapse
Affiliation(s)
- Nabila Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; International Institute of Translational Medicine, Jesionowa 11, 55-114, Malin, Wisznia Mała, Poland
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; International Institute of Translational Medicine, Jesionowa 11, 55-114, Malin, Wisznia Mała, Poland.
| |
Collapse
|
17
|
Vivero A, Ruz M, Rivera M, Miranda K, Sacristán C, Espinosa A, Codoceo J, Inostroza J, Vásquez K, Pérez Á, García-Díaz D, Arredondo M. Zinc Supplementation and Strength Exercise in Rats with Type 2 Diabetes: Akt and PTP1B Phosphorylation in Nonalcoholic Fatty Liver. Biol Trace Elem Res 2021; 199:2215-2224. [PMID: 32939643 DOI: 10.1007/s12011-020-02324-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes mellitus (T2D) is a metabolic disorder caused by chronic hyperglycemia due to a deficiency in the secretion and/or action of insulin. Zinc (Zn) supplementation and strength exercise increases insulin signaling. We evaluate the effect of Zn supplementation and strength exercise on insulin resistance in the liver of rats with diet-induced T2D through the study of phosphorylation of Akt and protein tyrosine phosphatase 1B (PTP1B). Rats were fed with a high-fat diet (HFD) for 18 weeks to induce T2D and then assigned in four experimental groups: HFD, HFD-Zn (Zn), HFD-strength exercise (Ex), and HFD-Zn/strength exercise (ZnEx) and treated during 12 weeks. Serum Zn, lipid profile, transaminases, glucose, and insulin were measured. In the liver with/without insulin stimuli, total and phosphorylated Akt (pAktSer473) and PTP1B (pPTP1BSer50) were determined by western blot. Hepatic steatosis was evaluated by histological staining with red oil and intrahepatic triglyceride (IHTG) content. There were no differences in biochemical and body-related variables. The ZnEx group showed a higher level of pAkt, both with/without insulin. The ZnEx group also showed higher levels of pPTP1B with respect to HFD and Zn groups. The ZnEx group had higher levels of pPTP1B than groups treated with insulin. Liver histology showed a better integrity and less IHTG in Ex and ZnEx with respect to the HFD group. The Ex and ZnEx groups had lower IHTG with respect to the HFD group. Our results showed that Zn supplementation and strength exercise together improved insulin signaling and attenuated nonalcoholic liver disease in a T2D rat model.
Collapse
Affiliation(s)
- Ariel Vivero
- Micronutrient Laboratory, Human Nutrition Unit, Institute of Nutrition and Food Technology, University of Chile, El Líbano 5524, Macul, Santiago, Chile
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Manuel Ruz
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Matías Rivera
- Micronutrient Laboratory, Human Nutrition Unit, Institute of Nutrition and Food Technology, University of Chile, El Líbano 5524, Macul, Santiago, Chile
| | - Karen Miranda
- Micronutrient Laboratory, Human Nutrition Unit, Institute of Nutrition and Food Technology, University of Chile, El Líbano 5524, Macul, Santiago, Chile
| | - Camila Sacristán
- Medical Technology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alejandra Espinosa
- Medical Technology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juana Codoceo
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jorge Inostroza
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Karla Vásquez
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Álvaro Pérez
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Diego García-Díaz
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Miguel Arredondo
- Micronutrient Laboratory, Human Nutrition Unit, Institute of Nutrition and Food Technology, University of Chile, El Líbano 5524, Macul, Santiago, Chile.
| |
Collapse
|
18
|
Liu YR, Wang JQ, Huang ZG, Chen RN, Cao X, Zhu DC, Yu HX, Wang XR, Zhou HY, Xia Q, Li J. Histone deacetylase‑2: A potential regulator and therapeutic target in liver disease (Review). Int J Mol Med 2021; 48:131. [PMID: 34013366 PMCID: PMC8136123 DOI: 10.3892/ijmm.2021.4964] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Histone acetyltransferases are responsible for histone acetylation, while histone deacetylases (HDACs) counteract histone acetylation. An unbalanced dynamic between histone acetylation and deacetylation may lead to aberrant chromatin landscape and chromosomal function. HDAC2, a member of class I HDAC family, serves a crucial role in the modulation of cell signaling, immune response and gene expression. HDAC2 has emerged as a promising therapeutic target for liver disease by regulating gene transcription, chromatin remodeling, signal transduction and nuclear reprogramming, thus receiving attention from researchers and clinicians. The present review introduces biological information of HDAC2 and its physiological and biochemical functions. Secondly, the functional roles of HDAC2 in liver disease are discussed in terms of hepatocyte apoptosis and proliferation, liver regeneration, hepatocellular carcinoma, liver fibrosis and non-alcoholic steatohepatitis. Moreover, abnormal expression of HDAC2 may be involved in the pathogenesis of liver disease, and its expression levels and pharmacological activity may represent potential biomarkers of liver disease. Finally, research on selective HDAC2 inhibitors and non-coding RNAs relevant to HDAC2 expression in liver disease is also reviewed. The aim of the present review was to improve understanding of the multifunctional role and potential regulatory mechanism of HDAC2 in liver disease.
Collapse
Affiliation(s)
- Ya-Ru Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Jie-Quan Wang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Zhao-Gang Huang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Ruo-Nan Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xi Cao
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Dong-Chun Zhu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Hai-Xia Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xiu-Rong Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Hai-Yun Zhou
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Jun Li
- The Key Laboratory of Anti‑inflammatory Immune Medicines, School of Pharmacy, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
19
|
Rocha S, Lucas M, Silva VLM, Gomes PMO, Silva AMS, Araújo AN, Aniceto N, Guedes RC, Corvo ML, Fernandes E, Freitas M. Pyrazoles as novel protein tyrosine phosphatase 1B (PTP1B) inhibitors: An in vitro and in silico study. Int J Biol Macromol 2021; 181:1171-1182. [PMID: 33857515 DOI: 10.1016/j.ijbiomac.2021.04.061] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 01/08/2023]
Abstract
Type 2 diabetes mellitus (DM) is a complex chronic disorder and a major global health problem. Insulin resistance is the primary detectable abnormality and the main characteristic feature in individuals with type 2 DM. Protein tyrosine phosphatase 1B (PTP1B) is a key negative regulator of the insulin signaling pathway, which dephosphorylates insulin receptor and insulin receptor substrates, suppressing the insulin signaling cascade. Therefore, the inhibition of PTP1B has become a potential strategy in the management of type 2 DM. In this study, a library of 22 pyrazoles was evaluated here for the first time against human PTP1B activity, using a microanalysis screening system. The results showed that 5-(2-hydroxyphenyl)-3-{2-[3-(4-nitrophenyl)-1,2,3,4-tetrahydronaphthyl]}-1-phenylpyrazole 20 and 3-(2-hydroxyphenyl)-5-{2-[3-(4-methoxyphenyl)]naphthyl}pyrazole 22 excelled as the most potent inhibitors of PTP1B, through noncompetitive inhibition mechanism. These findings suggest that the presence of additional benzene rings as functional groups in the pyrazole moiety increases the ability of pyrazoles to inhibit PTP1B. The most active compounds showed selectivity over the homologous T-cell protein tyrosine phosphatase (TCPTP). Molecular docking analyses were performed and revealed a particular contact signature involving residues like TYR46, ASP48, PHE182, TYR46, ALA217 and ILE219. This study represents a significant beginning for the design of novel PTP1B inhibitors.
Collapse
Affiliation(s)
- Sónia Rocha
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Mariana Lucas
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Vera L M Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pedro M O Gomes
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Artur M S Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Alberto N Araújo
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Natália Aniceto
- Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Rita C Guedes
- Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - M Luísa Corvo
- Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Eduarda Fernandes
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Marisa Freitas
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
20
|
Proença C, Ribeiro D, Freitas M, Carvalho F, Fernandes E. A comprehensive review on the antidiabetic activity of flavonoids targeting PTP1B and DPP-4: a structure-activity relationship analysis. Crit Rev Food Sci Nutr 2021; 62:4095-4151. [PMID: 33554619 DOI: 10.1080/10408398.2021.1872483] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Type 2 diabetes (T2D) is an expanding global health problem, resulting from defects in insulin secretion and/or insulin resistance. In the past few years, both protein tyrosine phosphatase 1B (PTP1B) and dipeptidyl peptidase-4 (DPP-4), as well as their role in T2D, have attracted the attention of the scientific community. PTP1B plays an important role in insulin resistance and is currently one of the most promising targets for the treatment of T2D, since no available PTP1B inhibitors were still approved. DPP-4 inhibitors are among the most recent agents used in the treatment of T2D (although its use has been associated with possible cardiovascular adverse events). The antidiabetic properties of flavonoids are well-recognized, and include inhibitory effects on the above enzymes, although hitherto not therapeutically explored. In the present study, a comprehensive review of the literature of both synthetic and natural isolated flavonoids as inhibitors of PTP1B and DPP-4 activities is made, including their type of inhibition and experimental conditions, and structure-activity relationship, covering a total of 351 compounds. We intend to provide the most favorable chemical features of flavonoids for the inhibition of PTP1B and DPP-4, gathering information for the future development of compounds with improved potential as T2D therapeutic agents.
Collapse
Affiliation(s)
- Carina Proença
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
21
|
Therapeutic Role of Protein Tyrosine Phosphatase 1B in Parkinson's Disease via Antineuroinflammation and Neuroprotection In Vitro and In Vivo. PARKINSONS DISEASE 2020; 2020:8814236. [PMID: 33456749 PMCID: PMC7787797 DOI: 10.1155/2020/8814236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/18/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD) is one of the most widespread neurodegenerative diseases. However, the currently available treatments could only relieve symptoms. Novel therapeutic targets are urgently needed. Several previous studies mentioned that protein tyrosine phosphatase 1B (PTP1B) acted as a negative regulator of the insulin signal pathway and played a significant role in the inflammation process. However, few studies have investigated the role of PTP1B in the central nervous system. Our study showed that suramin, an inhibitor of PTP1B, could improve neuronal damage. It could significantly attenuate the interferon-gamma-induced upregulation of proinflammatory cytokines, including inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). It enhanced M2 type microglia markers, such as arginase-1 and Ym-1 in BV2 murine microglial cells. PTP1B inhibition also reversed 6-hydroxydopamine- (6-OHDA-) induced downregulation of phospho-cAMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) in SH-SY5Y cells. Besides, we knocked down and overexpressed PTP1B in the SH-SY5Y cells to confirm its role in neuroprotection. We also verified the effect of suramin in the zebrafish PD model. Treatment with suramin could significantly reverse 6-OHDA-induced locomotor deficits and improved tyrosine hydroxylase (TH) via attenuating endoplasmic reticulum (ER) stress biomarkers. These results support that PTP1B could potentially regulate PD via antineuroinflammation and antiapoptotic pathways.
Collapse
|
22
|
Liu X, Yang Q, Nakamura Y. Inhibition of Drug Resistance Mechanisms Improves the Benzyl Isothiocyanate–Induced Anti-Proliferation in Human Colorectal Cancer Cells. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s40495-020-00227-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
23
|
Hou TY, Zhou Y, Zhu LS, Wang X, Pang P, Wang DQ, Liuyang ZY, Man H, Lu Y, Zhu LQ, Liu D. Correcting abnormalities in miR-124/PTPN1 signaling rescues tau pathology in Alzheimer's disease. J Neurochem 2020; 154:441-457. [PMID: 31951013 DOI: 10.1111/jnc.14961] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
MicroRNAs have been implicated in diverse physiological and pathological processes. We previously reported that aberrant microRNA-124 (miR-124)/non-receptor-type protein phosphatase 1 (PTPN1) signaling plays an important role in the synaptic disorders associated with Alzheimer's disease (AD). In this study, we further investigated the potential role of miR-124/PTPN1 in the tau pathology of AD. We first treated the mice with intra-hippocampal stereotactic injections. Then, we used quantitative real-time reverse transcription PCR (qRT-PCR) to detect the expression of microRNAs. Western blotting was used to measure the level of PTPN1, the level of tau protein, the phosphorylation of tau at AD-related sites, and alterations in the activity of glycogen synthase kinase 3β (GSK-3β) and protein phosphatase 2 (PP2A). Immunohistochemistry was also used to detect changes in tau phosphorylation levels at AD-related sites and somadendritic aggregation. Soluble and insoluble tau protein was separated by 70% formic acid (FA) extraction to examine tau solubility. Finally, behavioral experiments (including the Morris water maze, fear conditioning, and elevated plus maze) were performed to examine learning and memory ability and emotion-related behavior. We found that artificially replicating the abnormalities in miR-124/PTPN1 signaling induced AD-like tau pathology in the hippocampus of wild-type mice, including hyperphosphorylation at multiple sites, insolubility and somadendritic aggregation, as well as learning/memory deficits. We also found that disruption of miR-124/PTPN1 signaling was caused by the loss of RE1-silencing transcription factor protein, which can be initiated by Aβ insults or oxidative stress, as observed in the brains of P301S mice. Correcting the deregulation of miR-124/PTPN1 signaling rescued the tau pathology and learning/memory impairments in the P301S mice. We also found that miR-124/PTPN1 abnormalities induced activation of glycogen synthase kinase 3 (GSK-3) and inactivation of protein phosphatase 2A (PP2A) by promoting tyrosine phosphorylation, implicating an imbalance in tau kinase/phosphatase. Thus, targeting the miR-124/PTPN1 signaling pathway is a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Tong-Yao Hou
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yang Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ling-Shuang Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiong Wang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Pei Pang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ding-Qi Wang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhen-Yu Liuyang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hengye Man
- Department of Biology, Boston University, Boston, MA, USA
| | - Youming Lu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Dan Liu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China.,Department of Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
24
|
PTP1B promotes macrophage activation by regulating the NF-κB pathway in alcoholic liver injury. Toxicol Lett 2020; 319:11-21. [DOI: 10.1016/j.toxlet.2019.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023]
|
25
|
Kumar A, Rana D, Rana R, Bhatia R. Protein Tyrosine Phosphatase (PTP1B): A promising Drug Target Against Life-threatening Ailments. Curr Mol Pharmacol 2020; 13:17-30. [DOI: 10.2174/1874467212666190724150723] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/26/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022]
Abstract
Background:Protein tyrosine phosphatases are enzymes which help in the signal transduction in diabetes, obesity, cancer, liver diseases and neurodegenerative diseases. PTP1B is the main member of this enzyme from the protein extract of human placenta. In phosphate inhibitors development, significant progress has been made over the last 10 years. In early-stage clinical trials, few compounds have reached whereas in the later stage trials or registration, yet none have progressed. Many researchers investigate different ways to improve the pharmacological properties of PTP1B inhibitors.Objective:In the present review, authors have summarized various aspects related to the involvement of PTP1B in various types of signal transduction mechanisms and its prominent role in various diseases like cancer, liver diseases and diabetes mellitus.Conclusion:There are still certain challenges for the selection of PTP1B as a drug target. Therefore, continuous future efforts are required to explore this target for the development of PTP inhibitors to treat the prevailing diseases associated with it.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Pharmaceutical Analysis, Indo-Soviet Friendship College of Pharmacy (ISFCP), Moga-142001, India
| | - Divya Rana
- Department of Pharmaceutical Analysis, Indo-Soviet Friendship College of Pharmacy (ISFCP), Moga-142001, India
| | - Rajat Rana
- Department of Pharmaceutical Analysis, Indo-Soviet Friendship College of Pharmacy (ISFCP), Moga-142001, India
| | - Rohit Bhatia
- Department of Pharmaceutical Analysis, Indo-Soviet Friendship College of Pharmacy (ISFCP), Moga-142001, India
| |
Collapse
|
26
|
Feng CW, Chen NF, Chan TF, Chen WF. Therapeutic Role of Protein Tyrosine Phosphatase 1B in Parkinson's Disease via Antineuroinflammation and Neuroprotection In Vitro and In Vivo. PARKINSON'S DISEASE 2020. [PMID: 33456749 DOI: 10.1155/2020/8814236.ecollection2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Parkinson's disease (PD) is one of the most widespread neurodegenerative diseases. However, the currently available treatments could only relieve symptoms. Novel therapeutic targets are urgently needed. Several previous studies mentioned that protein tyrosine phosphatase 1B (PTP1B) acted as a negative regulator of the insulin signal pathway and played a significant role in the inflammation process. However, few studies have investigated the role of PTP1B in the central nervous system. Our study showed that suramin, an inhibitor of PTP1B, could improve neuronal damage. It could significantly attenuate the interferon-gamma-induced upregulation of proinflammatory cytokines, including inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). It enhanced M2 type microglia markers, such as arginase-1 and Ym-1 in BV2 murine microglial cells. PTP1B inhibition also reversed 6-hydroxydopamine- (6-OHDA-) induced downregulation of phospho-cAMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) in SH-SY5Y cells. Besides, we knocked down and overexpressed PTP1B in the SH-SY5Y cells to confirm its role in neuroprotection. We also verified the effect of suramin in the zebrafish PD model. Treatment with suramin could significantly reverse 6-OHDA-induced locomotor deficits and improved tyrosine hydroxylase (TH) via attenuating endoplasmic reticulum (ER) stress biomarkers. These results support that PTP1B could potentially regulate PD via antineuroinflammation and antiapoptotic pathways.
Collapse
Affiliation(s)
- Chien-Wei Feng
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807377, Taiwan
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Te-Fu Chan
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807377, Taiwan
| | - Wu-Fu Chen
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Neurosurgery, Xiamen Chang Gung Hospital, Xiamen, Fujian, China
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
27
|
Protein tyrosine phosphatase 1b deficiency protects against hepatic fibrosis by modulating nadph oxidases. Redox Biol 2019; 26:101263. [PMID: 31299613 PMCID: PMC6624458 DOI: 10.1016/j.redox.2019.101263] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/30/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023] Open
Abstract
Inflammation is typically associated with the development of fibrosis, cirrhosis and hepatocellular carcinoma. The key role of protein tyrosine phosphatase 1B (PTP1B) in inflammatory responses has focused this study in understanding its implication in liver fibrosis. Here we show that hepatic PTP1B mRNA expression increased after bile duct ligation (BDL), while BDL-induced liver fibrosis was markedly reduced in mice lacking Ptpn1 (PTP1B−/−) as assessed by decreased collagen deposition and α-smooth muscle actin (α-SMA) expression. PTP1B−/− mice also showed a significant increase in mRNA levels of key markers of monocytes recruitment (Cd68, Adgre1 and Ccl2) compared to their wild-type (PTP1B+/+) littermates at early stages of injury after BDL. Interestingly, the lack of PTP1B strongly increased the NADPH oxidase (NOX) subunits Nox1/Nox4 ratio and downregulated Cybb expression after BDL, revealing a pro-survival pattern of NADPH oxidase induction in response to liver injury. Chimeric mice generated by transplantation of PTP1B−/− bone marrow (BM) into irradiated PTP1B+/+ mice revealed similar hepatic expression profile of NOX subunits than PTP1B−/− mice while these animals did not show differences in infiltration of myeloid cells at 7 days post-BDL, suggesting that PTP1B deletion in other liver cells is necessary for boosting the early inflammatory response to the BDL. PTP1B−/− BM transplantation into PTP1B+/+ mice also led to a blockade of TGF-β and α-SMA induction after BDL. In vitro experiments demonstrated that deficiency of PTP1B in hepatocytes protects against bile acid-induced apoptosis and abrogates hepatic stellate cells (HSC) activation, an effect ameliorated by NOX1 inhibition. In conclusion, our results have revealed that the lack of PTP1B switches NOX expression pattern in response to liver injury after BDL and reduces HSC activation and liver fibrosis. PTP1B deficiency in mice ameliorates liver damage induced by cholestasis. The increased NOX1/NOX4 ratio in livers from PTP1B-/- mice was associated with protection against BDL-induced fibrosis. The lack of PTP1B exacerbates macrophage recruitment upon BDL which is dispensable for ameliorating cholestatic liver damage. Resistance of PTP1B-/- hepatocytes against bile acid-induced apoptosis protects from HSC activation in a NOX1-dependent manner.
Collapse
|
28
|
Xue W, Tian J, Wang XS, Xia J, Wu S. Discovery of potent PTP1B inhibitors via structure-based drug design, synthesis and in vitro bioassay of Norathyriol derivatives. Bioorg Chem 2019; 86:224-234. [PMID: 30716620 DOI: 10.1016/j.bioorg.2019.01.059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/11/2019] [Accepted: 01/27/2019] [Indexed: 12/11/2022]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) has recently been identified as a potential target of Norathyriol. Unfortunately, Norathyriol is not a potent PTP1B inhibitor, which somewhat hinders its further application. Based on the fact that no study on the relationship of chemical structure and PTP1B inhibitory activity of Norathyriol has been reported so far, we attempted to perform structural optimization so as to improve the potency for PTP1B. Via structure-based drug design (SBDD), a rational strategy based on the binding mode of Norathyriol to PTP1B, we designed 26 derivatives with substitutions at the four phenolic hydroxyl groups of Norathyriol. By chemical synthesis and in vitro bioassay, we identified seven PTP1B inhibitors that were more potent than Norathyriol, of which XWJ24 showed the highest potency (IC50: 0.6 μM). We also found out that XWJ24 was a competitive inhibitor and showed the 4.5-fold selectivity over its close homolog, TC-PTP. Through molecular docking of XWJ24 against PTP1B, we highlighted the essential role of its hydrogen bond with Asp181 for PTP1B inhibition and identified a potential halogen bond with Asp48 that was not observed for Norathyriol. The current data indicate that our SBDD strategy is effective to discover potent PTP1B-targeted Norathyriol derivatives, and XWJ24 is a promising lead compound for further development.
Collapse
Affiliation(s)
- Wenjie Xue
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jinlong Tian
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiang Simon Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington DC 20059, USA
| | - Jie Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Song Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
29
|
Chu MJ, Tang XL, Han X, Li T, Luo XC, Jiang MM, van Ofwegen L, Luo LZ, Zhang G, Li PL, Li GQ. Metabolites from the Paracel Islands Soft Coral Sinularia cf. molesta. Mar Drugs 2018; 16:md16120517. [PMID: 30572615 PMCID: PMC6317055 DOI: 10.3390/md16120517] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 12/13/2022] Open
Abstract
Five new oxygenated sesquiterpenes, molestins A–D (1, 3–5) and epi-gibberodione (2), three new cyclopentenone derivatives, ent-sinulolides C, D, and F ((+)-9–(+)-11), one new butenolide derivative, ent-sinulolide H ((+)-13), and one new cembranolide, molestin E (14), together with 14 known related metabolites (6–8, (–)-9–(–)-11, (±)-12, (–)-13, 15–19) were isolated from the Paracel Islands soft coral Sinularia cf. molesta. The structures and absolute configurations were elucidated based on comprehensive spectroscopic analyses, quantum chemical calculations, and comparison with the literature data. Compound 5 is the first example of a norsesquiterpene with a de-isopropyl guaiane skeleton isolated from the genus Sinularia. Molestin E (14) exhibited cytotoxicities against HeLa and HCT-116 cell lines with IC50 values of 5.26 and 8.37 μM, respectively. Compounds 4, 5, and 8 showed significant inhibitory activities against protein tyrosine phosphatase 1B (PTP1B) with IC50 values of 218, 344, and 1.24 μM, respectively.
Collapse
Affiliation(s)
- Mei-Jun Chu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao 266101, China.
| | - Xu-Li Tang
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266100, China.
| | - Xiao Han
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Laboratory of Marine Drugs and Biological Products, National Laboratory for Marine Science and Technology, Qingdao 266235, China.
| | - Tao Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Laboratory of Marine Drugs and Biological Products, National Laboratory for Marine Science and Technology, Qingdao 266235, China.
| | - Xiang-Chao Luo
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Laboratory of Marine Drugs and Biological Products, National Laboratory for Marine Science and Technology, Qingdao 266235, China.
| | - Ming-Ming Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Laboratory of Marine Drugs and Biological Products, National Laboratory for Marine Science and Technology, Qingdao 266235, China.
| | - Leen van Ofwegen
- Nationaal Natuurhistorisch Museum, 2300 RA Leiden, The Netherlands.
| | - Lian-Zhong Luo
- Xiamen Key Laboratory of Marine Medicinal Natural Products Resources, Xiamen Medical College, Xiamen 361023, China.
| | - Gang Zhang
- Xiamen Key Laboratory of Marine Medicinal Natural Products Resources, Xiamen Medical College, Xiamen 361023, China.
| | - Ping-Lin Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Laboratory of Marine Drugs and Biological Products, National Laboratory for Marine Science and Technology, Qingdao 266235, China.
| | - Guo-Qiang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Laboratory of Marine Drugs and Biological Products, National Laboratory for Marine Science and Technology, Qingdao 266235, China.
| |
Collapse
|
30
|
Mojena M, Pimentel-Santillana M, Povo-Retana A, Fernández-García V, González-Ramos S, Rada P, Tejedor A, Rico D, Martín-Sanz P, Valverde AM, Boscá L. Protection against gamma-radiation injury by protein tyrosine phosphatase 1B. Redox Biol 2018; 17:213-223. [PMID: 29705509 PMCID: PMC6006913 DOI: 10.1016/j.redox.2018.04.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/13/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is widely expressed in mammalian tissues, in particular in immune cells, and plays a pleiotropic role in dephosphorylating many substrates. Moreover, PTP1B expression is enhanced in response to pro-inflammatory stimuli and to different cell stressors. Taking advantage of the use of mice deficient in PTP1B we have investigated the effect of γ-radiation in these animals and found enhanced lethality and decreased respiratory exchange ratio vs. the corresponding wild type animals. Using bone-marrow derived macrophages and mouse embryonic fibroblasts (MEFs) from wild-type and PTP1B-deficient mice, we observed a differential response to various cell stressors. PTP1B-deficient macrophages exhibited an enhanced response to γ-radiation, UV-light, LPS and S-nitroso-glutathione. Macrophages exposed to γ-radiation show DNA damage and fragmentation, increased ROS production, a lack in GSH elevation and enhanced acidic β-galactosidase activity. Interestingly, these differences were not observed in MEFs. Differential gene expression analysis of WT and KO macrophages revealed that the main pathways affected after irradiation were an up-regulation of protein secretion, TGF-β signaling and angiogenesis among other, and downregulation of Myc targets and Hedgehog signaling. These results demonstrate a key role for PTP1B in the protection against the cytotoxicity of irradiation in intact animal and in macrophages, which might be therapeutically relevant.
Collapse
Affiliation(s)
- Marina Mojena
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | - María Pimentel-Santillana
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | - Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | - Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), y Hepáticas y Digestivas (CIBEREHD), ISCIII, Spain
| | - Patricia Rada
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Alberto Tejedor
- Hospital General Universitario Gregorio Marañón, Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Daniel Rico
- Institute of Cellular Medicine, Newcastle University, United Kingdom
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), y Hepáticas y Digestivas (CIBEREHD), ISCIII, Spain
| | - Angela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain; Unidad Asociada IIBM-ULPGC, Universidad de las Palmas de Gran Canaria (ULPGC), Spain.
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), y Hepáticas y Digestivas (CIBEREHD), ISCIII, Spain; Unidad Asociada IIBM-ULPGC, Universidad de las Palmas de Gran Canaria (ULPGC), Spain.
| |
Collapse
|
31
|
Proença C, Freitas M, Ribeiro D, Sousa JLC, Carvalho F, Silva AMS, Fernandes PA, Fernandes E. Inhibition of protein tyrosine phosphatase 1B by flavonoids: A structure - activity relationship study. Food Chem Toxicol 2017; 111:474-481. [PMID: 29175190 DOI: 10.1016/j.fct.2017.11.039] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/05/2017] [Accepted: 11/19/2017] [Indexed: 12/23/2022]
Abstract
The classical non-transmembrane protein tyrosine phosphatase 1B (PTP1B) has emerged as a key negative regulator of insulin signaling pathways that leads to insulin resistance, turning this enzyme a promising therapeutic target in the management of type 2 diabetes mellitus (T2DM). In the present work, the in vitro inhibitory activity of a panel of structurally related flavonoids, for recombinant human PTP1B was studied and the type of inhibition of the most active compounds further evaluated. The majority of the studied flavonoids was tested in this work for the first time, including flavonoid C13, which was the most potent inhibitor. It was observed that the ability to inhibit PTP1B depends on the nature, position and number of substituents in the flavonoid structure, as the presence of both 7- and 8-OBn groups in the A ring, together with the presence of both 3' and 4'-OMe groups in the B ring and the 3-OH group in the C ring; these substituents increase the flavonoids' ability to inhibit PTP1B. In conclusion, some of the tested flavonoids seem to be promising PTP1B inhibitors and potential effective agents in the management of T2DM, by increasing insulin sensitivity.
Collapse
Affiliation(s)
- Carina Proença
- UCIBIO, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Marisa Freitas
- UCIBIO, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniela Ribeiro
- UCIBIO, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana L C Sousa
- Department of Chemistry & QOPNA, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Artur M S Silva
- Department of Chemistry & QOPNA, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pedro A Fernandes
- UCIBIO, REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Eduarda Fernandes
- UCIBIO, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
32
|
Sun W, Sun J, Zhang B, Xing Y, Yu X, Li X, Xiu Z, Dong Y. Baicalein improves insulin resistance via regulating SOCS3 and enhances the effect of acarbose on diabetes prevention. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
33
|
Sun W, Zhuang C, Li X, Zhang B, Lu X, Zheng Z, Dong Y. Varic acid analogues from fungus as PTP1B inhibitors: Biological evaluation and structure-activity relationships. Bioorg Med Chem Lett 2017. [PMID: 28642102 DOI: 10.1016/j.bmcl.2017.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) inhibitors as potential therapies for diabetes and obesity have attracted much attention in recent years. Six varic acid analogues were isolated from two strains of fungi and evaluated for PTP1B inhibition activities. The structure-activity relationships were also characterized and predicted by molecular modeling. Further kinetic studies indicated the reversible and competitive inhibition manner of varic acid analogues. Trivaric acid showed insulin-sensitizing effect not only in vitro but also in vivo, representing a promising lead compound for further optimization.
Collapse
Affiliation(s)
- Wenlong Sun
- School of Life Science, Biotechnology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Chunlin Zhuang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.
| | - Xia Li
- School of Life Science, Biotechnology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Bowei Zhang
- School of Life Science, Biotechnology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xinhua Lu
- New Drug Research and Development Center, North China Pharmaceutical Group Corporation and National Microbial Medicine Engineering and Research Center, Shijiazhuang, Hebei 050015, China.
| | - Zhihui Zheng
- New Drug Research and Development Center, North China Pharmaceutical Group Corporation and National Microbial Medicine Engineering and Research Center, Shijiazhuang, Hebei 050015, China
| | - Yuesheng Dong
- School of Life Science, Biotechnology, Dalian University of Technology, Dalian, Liaoning 116024, China.
| |
Collapse
|
34
|
Gajęcka M, Przybylska-Gornowicz B, Zakłos-Szyda M, Dąbrowski M, Michalczuk L, Koziołkiewicz M, Babuchowski A, Zielonka Ł, Lewczuk B, Gajęcki MT. The influence of a natural triterpene preparation on the gastrointestinal tract of gilts with streptozocin-induced diabetes and on cell metabolic activity. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.03.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
35
|
Le DD, Nguyen DH, Zhao BT, Seong SH, Choi JS, Kim SK, Kim JA, Min BS, Woo MH. PTP1B inhibitors from Selaginella tamariscina (Beauv.) Spring and their kinetic properties and molecular docking simulation. Bioorg Chem 2017; 72:273-281. [DOI: 10.1016/j.bioorg.2017.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/03/2017] [Accepted: 05/01/2017] [Indexed: 02/05/2023]
|
36
|
Seong SH, Roy A, Jung HA, Jung HJ, Choi JS. Protein tyrosine phosphatase 1B and α-glucosidase inhibitory activities of Pueraria lobata root and its constituents. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:706-716. [PMID: 27769948 DOI: 10.1016/j.jep.2016.10.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 08/29/2016] [Accepted: 10/03/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pueraria lobata root was used to treat wasting-thirst regarded as diabetes mellitus and was included in the composition of Okcheonsan, which is prescribed for thirst-waste in traditional Chinese medicine. AIM OF THE STUDY The objective of this study was to evaluate the anti-diabetic potential of the root of Pueraria lobata and its constituents via protein tyrosine phosphatase 1B (PTP1B) and α-glucosidase inhibitory activities. MATERIALS AND METHODS In this study, anti-diabetic activities of the 70% ethanolic (EtOH) extract from P. lobata roots and its solvent soluble fractions with the isolated compounds were investigated by evaluating in vitro PTP1B and α-glucosidase inhibitory activities. We also examined the potentials of active compounds as PTP1B and α-glucosidase inhibitors via enzyme kinetics and in silico molecular docking simulation between the enzymes and active compounds. RESULTS Triterpenoids lupeol and lupenone were potent PTP1B inhibitors with IC50 values of 38.89±0.17 and 15.11±1.23μM. Kinetic study using the Lineweaver-Burk and Dixon plots demonstrated that these compounds showed a noncompetitive-type inhibition against PTP1B with respective Ki values of 13.88μM and 21.24μM. In addition, molecular docking simulation showed lupeol and lupenone has negative binding energy values of -8.03 and -8.56kcal/mol. Considering the α-glucosidase inhibitory potential, daidzein, genistein, and calycosin exhibited the most potent α-glucosidase inhibition with IC50 values of 8.58±0.94, 2.37±0.52 and 6.84±1.58μM, respectively. Kinetic study demonstrated that these 3 compounds showed a noncompetitive-type inhibition against α-glucosidase with respective Ki values of 17.64μM, 5.03μM and 13.83μM. Moreover, molecular docking simulation showed daidzein, genistein and calycosin has more lower binding energy (-7.16kcal/mol, -7.42kcal/mol and -7.31kcal/mol) with higher binding affinity and tight binding capacity in the molecular docking studies than standard ligand α-D-glucose (-6.74kcal/mol). CONCLUSION Our results of the present study clearly demonstrate the potential of P. lobata extract and its constituents to inhibit PTP1B and α-glucosidase, contributing to the development of therapeutic or preventive agents that can be used in the treatment of diabetes.
Collapse
Affiliation(s)
- Su Hui Seong
- Department of Food and Life Science, Pukyong National University, Busan 608-737, Republic of Korea
| | - Anupom Roy
- Department of Food and Life Science, Pukyong National University, Busan 608-737, Republic of Korea
| | - Hyun Ah Jung
- Department of Food Science and Human Nutrition, Chonbuk National University, Jeonju 561-756, Republic of Korea.
| | - Hee Jin Jung
- Department of Food and Life Science, Pukyong National University, Busan 608-737, Republic of Korea
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan 608-737, Republic of Korea.
| |
Collapse
|
37
|
Sun W, Zhang B, Zheng H, Zhuang C, Li X, Lu X, Quan C, Dong Y, Zheng Z, Xiu Z. Trivaric acid, a new inhibitor of PTP1b with potent beneficial effect on diabetes. Life Sci 2016; 169:52-64. [PMID: 27871946 DOI: 10.1016/j.lfs.2016.11.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/02/2016] [Accepted: 11/17/2016] [Indexed: 11/30/2022]
Abstract
AIM To screen a potential PTP1b inhibitor from the microbial origin-based compound library and to investigate the potential anti-diabetic effects of the inhibitor in vivo and determine its primary anti-diabetic mechanism in vitro and in silico. METHODS PTP1b inhibitory activity was measured using recombination protein as the enzyme and p-NPP as the substrate. The binding of the inhibitor to PTP1b was analysed by docking in silico and confirmed by ITC experiments. The intracellular signalling pathway was detected by Western blot analysis in HepG2 cells. The anti-diabetic effects were evaluated using a diabetic mice model in vivo. RESULTS Among 545 microbial origin-based pure compounds tested, trivaric acid, a tridepside, was selected as a PTP1B inhibitor exhibiting strong inhibitory activity with an IC50 of 173nM. Docking and ITC studies showed that trivaric acid was able to spontaneously bind to PTP1b and may inhibit PTP1b by blocking the catalytic domain of the phosphatase. Trivaric acid also enhanced the ability of insulin to stimulate the IR/IRS/Akt/GLUT2 pathway and increase the glucose consumption in HepG2 cells. In diabetic mice, trivaric acid that had been encapsulated into Eudrgit L100-5.5 showed significant anti-diabetic effects, improving insulin resistance, leptin resistance and lipid profile and weight control at doses of 5mg/kg and 50mg/kg. SIGNIFICANCE Trivaric acid is a potential lead compound in the search for anti-diabetic agents targeting PTP1b.
Collapse
Affiliation(s)
- Wenlong Sun
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Bowei Zhang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Haizhou Zheng
- New Drug Research and Development Center, North China Pharmaceutical Group Corporation, National Microbial Medicine Engineering and Research Center, Shijiazhuang 050015, China
| | - Chunlin Zhuang
- Research Center for Marine Drugs, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Xia Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Xinhua Lu
- New Drug Research and Development Center, North China Pharmaceutical Group Corporation, National Microbial Medicine Engineering and Research Center, Shijiazhuang 050015, China
| | - Chunshan Quan
- Key Laboratory of Biotechnology and Bioresources Utilization, Dalian Minzu University, Dalian 116024, Liaoning, China
| | - Yuesheng Dong
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China.
| | - Zhihui Zheng
- New Drug Research and Development Center, North China Pharmaceutical Group Corporation, National Microbial Medicine Engineering and Research Center, Shijiazhuang 050015, China.
| | - Zhilong Xiu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| |
Collapse
|
38
|
Cocoa bean (Theobroma cacao L.) phenolic extracts as PTP1B inhibitors, hepatic HepG2 and pancreatic β-TC3 cell cytoprotective agents and their influence on oxidative stress in rats. Food Res Int 2016. [DOI: 10.1016/j.foodres.2016.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
39
|
Rines AK, Sharabi K, Tavares CDJ, Puigserver P. Targeting hepatic glucose metabolism in the treatment of type 2 diabetes. Nat Rev Drug Discov 2016; 15:786-804. [PMID: 27516169 DOI: 10.1038/nrd.2016.151] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus is characterized by the dysregulation of glucose homeostasis, resulting in hyperglycaemia. Although current diabetes treatments have exhibited some success in lowering blood glucose levels, their effect is not always sustained and their use may be associated with undesirable side effects, such as hypoglycaemia. Novel antidiabetic drugs, which may be used in combination with existing therapies, are therefore needed. The potential of specifically targeting the liver to normalize blood glucose levels has not been fully exploited. Here, we review the molecular mechanisms controlling hepatic gluconeogenesis and glycogen storage, and assess the prospect of therapeutically targeting associated pathways to treat type 2 diabetes.
Collapse
Affiliation(s)
- Amy K Rines
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Clint D J Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
40
|
Chen PJ, Cai SP, Yang Y, Li WX, Huang C, Meng XM, Li J. PTP1B confers liver fibrosis by regulating the activation of hepatic stellate cells. Toxicol Appl Pharmacol 2016; 292:8-18. [DOI: 10.1016/j.taap.2015.12.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 01/21/2023]
|