1
|
Liang Q, Huang Z, Wang D, Xin F, Xu J, He J, Luo R, Zeng Y, Ma Y. Study on the role of miR-10b-3p as SASP in exosomes of premature senescent hepatocytes induced by hexavalent chromium. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118333. [PMID: 40373709 DOI: 10.1016/j.ecoenv.2025.118333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
The exact mechanisms through which chronic, low-concentration exposure to Cr(VI) facilitates the development of related pathological conditions remain to be fully elucidated. Senescence-associated secretory phenotype (SASP) exhibits a bidirectional regulatory function in biological processes. Consequently, it is essential to identify the formulation and functional characteristics of the SASP released by Cr(VI)-triggered senescent L02 hepatocytes (S-L02). In this study, exosomes were extracted from the conditioned media of both normal L02 cells and their senescent counterparts (Senescent L02, S-L02 cells). Among the miRNAs identified in the exosomes, miR-10b-3p was found to be the most abundantly expressed in the exosomes derived from S-L02 cells. As part of the SASP, miR-10b-3p was shown to suppress the proliferation of both L02 and S-L02 cells. Simultaneously, it promoted the growth, migration, and invasive capabilities of hepatocellular carcinoma (HCC) cells. The next mechanistic analysis showed that miR-10b-3p reduces the regulatory influence of the protein PHLPP2 on Akt by downregulating its target gene, PHLPP2. This suppression led to lower levels of p27, FOXO3a and p21, thereby enhancing the proliferation of HCC cells by relieving the negative regulatory mechanisms of the cell cycle. This research offers significant understanding into the oncogenic pathways induced by Cr(VI), and provides laboratory evidence for mechanistic studies targeting hepatic carcinoma associated with Cr(VI) exposure.
Collapse
Affiliation(s)
- Qi Liang
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zidi Huang
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Di Wang
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Fei Xin
- Dalian Health Development Center, Dalian 116011, China
| | - Jiaqi Xu
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Jiajia He
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Runnan Luo
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Yuan Zeng
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China.
| | - Yu Ma
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
2
|
Mohammed Abdul KS, Han K, Guerrero AB, Wilson CN, Kulkarni A, Purcell NH. Increased PHLPP1 expression through ERK-4E-BP1 signaling axis drives nicotine induced oxidative stress related damage of cardiomyocytes. J Mol Cell Cardiol 2024; 193:100-112. [PMID: 38851627 DOI: 10.1016/j.yjmcc.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Nicotine, a key constituent of tobacco/electronic cigarettes causes cardiovascular injury and mortality. Nicotine is known to induce oxidative stress and mitochondrial dysfunction in cardiomyocytes leading to cell death. However, the underlying mechanisms remain unclear. Pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) is a member of metal-dependent protein phosphatase (PPM) family and is known to dephosphorylate several AGC family kinases and thereby regulate a diverse set of cellular functions including cell growth, survival, and death. Our lab has previously demonstrated that PHLPP1 removal reduced cardiomyocyte death and cardiac dysfunction following injury. Here, we present a novel finding that nicotine exposure significantly increased PHLPP1 protein expression in the adolescent rodent heart. Building upon our in vivo finding, we determined the mechanism of PHLPP1 expression in cardiomyocytes. Nicotine significantly increased PHLPP1 protein expression without altering PHLPP2 in cardiomyocytes. In cardiomyocytes, nicotine significantly increased NADPH oxidase 4 (NOX4), which coincided with increased reactive oxygen species (ROS) and increased cardiomyocyte apoptosis which were dependent on PHLPP1 expression. PHLPP1 expression was both necessary and sufficient for nicotine induced mitochondrial dysfunction. Mechanistically, nicotine activated extracellular signal-regulated protein kinases (ERK1/2) and subsequent eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) to increase PHLPP1 protein expression. Inhibition of protein synthesis with cycloheximide (CHX) and 4EGI-1 abolished nicotine induced PHLPP1 protein expression. Moreover, inhibition of ERK1/2 activity by U0126 significantly blocked nicotine induced PHLPP1 expression. Overall, this study reveals a novel mechanism by which nicotine regulates PHLPP1 expression through ERK-4E-BP1 signaling axis to drive cardiomyocyte injury.
Collapse
Affiliation(s)
| | - Kimin Han
- Cardiovascular Signaling Division, Huntington Medical Research Institutes, Pasadena, California, USA
| | - Alyssa B Guerrero
- Cardiovascular Signaling Division, Huntington Medical Research Institutes, Pasadena, California, USA
| | - Cekia N Wilson
- Cardiovascular Signaling Division, Huntington Medical Research Institutes, Pasadena, California, USA
| | - Amogh Kulkarni
- Cardiovascular Signaling Division, Huntington Medical Research Institutes, Pasadena, California, USA
| | - Nicole H Purcell
- Cardiovascular Signaling Division, Huntington Medical Research Institutes, Pasadena, California, USA; Cardiovascular Division, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
3
|
Liu X, Liu Y, Chen L, Zhang Z, Cui L, Wei T. Loss of pleckstrin homology domain and leucine-rich repeat protein phosphatase 2 has protective effects on high glucose-injured retinal ganglion cells via the effect on the Akt-GSK-3β-Nrf2 pathway. Inflamm Res 2023; 72:373-385. [PMID: 36562794 DOI: 10.1007/s00011-022-01680-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Pleckstrin homology domain and leucine-rich repeat protein phosphatase 2 (PHLPP2) is linked to various pathological states. However, whether PHLPP2 mediates diabetic retinopathy is unaddressed. This work explored the biological function of PHLPP2 in modulating high glucose (HG)-elicited damage of retinal ganglion cells (RGCs), an in vitro model for studying diabetic retinopathy. METHODS Mouse RGCs were treated with HG to establish a cell model. PHLPP2 was silenced by transfecting specific shRNAs targeting PHLPP2. RT-qPCR, immunoblotting, CCK-8 assay, flow cytometry, TUNEL assay, and ELISA were carried out. RESULTS Significant increases in PHLPP2 levels were observed in cultured RGCs exposed to HG. The severe damages evoked by HG to RGCs were remarkably weakened in PHLPP2-silenced RGCs, including improved cell survival, attenuated cell apoptosis, repressed oxidative stress, and prohibited proinflammatory response. The silencing of PHLPP2 strengthened the activation of Nrf2 in HG-treated RGCs via modulation of the Akt-GSK-3β axis. Interruption of the Akt-GSK-3β axis reversed PHLPP2-silencing-elicited Nrf2 activation. The protective effects of PHLPP2 silencing on HG-induced injury of RGCs were diminished by Nrf2 inhibition. CONCLUSIONS The loss of PHLPP2 was beneficial for HG-injured RGCs through the effect on the Akt-GSK-3β-Nrf2 pathway. This work suggests a possible role of PHLPP2 in diabetic retinopathy.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| | - Yong Liu
- The Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta Road, Xi'an, 710061, Shaanxi, China.
| | - Li Chen
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Zhichao Zhang
- The Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Lijun Cui
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Ting Wei
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| |
Collapse
|
4
|
Balamurugan K, Chandra K, Sai Latha S, Swathi M, Joshi MB, Misra P, Parsa KVL. PHLPPs: Emerging players in metabolic disorders. Drug Discov Today 2022; 27:103317. [PMID: 35835313 DOI: 10.1016/j.drudis.2022.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/21/2022] [Accepted: 07/07/2022] [Indexed: 12/15/2022]
Abstract
That reversible protein phosphorylation by kinases and phosphatases occurs in metabolic disorders is well known. Various studies have revealed that a multi-faceted and tightly regulated phosphatase, pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP)-1/2 displays robust effects in cardioprotection, ischaemia/reperfusion (I/R), and vascular remodelling. PHLPP1 promotes foamy macrophage development through ChREBP/AMPK-dependent pathways. Adipocyte-specific loss of PHLPP2 reduces adiposity, improves glucose tolerance,and attenuates fatty liver via the PHLPP2-HSL-PPARα axis. Discoveries of PHLPP1-mediated insulin resistance and pancreatic β cell death via the PHLPP1/2-Mst1-mTORC1 triangular loop have shed light on its significance in diabetology. PHLPP1 downregulation attenuates diabetic cardiomyopathy (DCM) by restoring PI3K-Akt-mTOR signalling. In this review, we summarise the functional role of, and cellular signalling mediated by, PHLPPs in metabolic tissues and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Keerthana Balamurugan
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India; Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Kanika Chandra
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India; Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - S Sai Latha
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - M Swathi
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Parimal Misra
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - Kishore V L Parsa
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India.
| |
Collapse
|
5
|
Transcription factor SNAI2 exerts pro-tumorigenic effects on glioma stem cells via PHLPP2-mediated Akt pathway. Cell Death Dis 2022; 13:516. [PMID: 35654777 PMCID: PMC9163135 DOI: 10.1038/s41419-021-04481-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 12/01/2021] [Accepted: 12/17/2021] [Indexed: 01/21/2023]
Abstract
The current study aimed to investigate the effects associated with SNAI2 on the proliferation of glioma stem cells (GSCs) to elucidate its underlying molecular mechanism in the development of glioma. The expression of Snail family transcriptional repressor 2 (SNAI2) in glioma tissues was initially predicted via bioinformatics analysis and subsequently confirmed by reverse transcription quantitative polymerase chain reaction (RT-qPCR), which revealed that SNAI2 was highly expressed in glioma tissues as well as GSCs, with an inverse correlation with overall glioma patient survival detected. Loss- and gain- of-function assays were performed to determine the roles of SNAI2 and pleckstrin homology domain and leucine rich repeat protein phosphatase 2 (PHLPP2) on GSC viability, proliferation and apoptosis. Data were obtained indicating that SNAI2 promoted the proliferation of GSCs, while overexpressed PHLPP2 brought about a contrasting trend. As detected by chromatin immunoprecipitation, RT-qPCR and agarose gel electrophoresis, SNAI2 bound to the promoter region of PHLPP2 and repressed the transcription of PHLPP2 while SNAI2 was found to inhibit PHLPP2 resulting in activation of the Akt pathway. Finally, the roles of SNAI2 and PHLPP2 were verified in glioma growth in nude mice xenografted with tumor. Taken together, the key findings of the present study suggest that SNAI2 may promote the proliferation of GSCs through activation of the Akt pathway by downregulating PHLPP2.
Collapse
|
6
|
de Lucia C, Grisanti LA, Borghetti G, Piedepalumbo M, Ibetti J, Lucchese AM, Barr EW, Roy R, Okyere AD, Murphy HC, Gao E, Rengo G, Houser SR, Tilley DG, Koch WJ. G protein-coupled receptor kinase 5 (GRK5) contributes to impaired cardiac function and immune cell recruitment in post-ischemic heart failure. Cardiovasc Res 2022; 118:169-183. [PMID: 33560342 PMCID: PMC8752360 DOI: 10.1093/cvr/cvab044] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/15/2020] [Accepted: 02/05/2021] [Indexed: 12/25/2022] Open
Abstract
AIMS Myocardial infarction (MI) is the most common cause of heart failure (HF) worldwide. G protein-coupled receptor kinase 5 (GRK5) is upregulated in failing human myocardium and promotes maladaptive cardiac hypertrophy in animal models. However, the role of GRK5 in ischemic heart disease is still unknown. In this study, we evaluated whether myocardial GRK5 plays a critical role post-MI in mice and included the examination of specific cardiac immune and inflammatory responses. METHODS AND RESULTS Cardiomyocyte-specific GRK5 overexpressing transgenic mice (TgGRK5) and non-transgenic littermate control (NLC) mice as well as cardiomyocyte-specific GRK5 knockout mice (GRK5cKO) and wild type (WT) were subjected to MI and, functional as well as structural changes together with outcomes were studied. TgGRK5 post-MI mice showed decreased cardiac function, augmented left ventricular dimension and decreased survival rate compared to NLC post-MI mice. Cardiac hypertrophy and fibrosis as well as fetal gene expression were increased post-MI in TgGRK5 compared to NLC mice. In TgGRK5 mice, GRK5 elevation produced immuno-regulators that contributed to the elevated and long-lasting leukocyte recruitment into the injured heart and ultimately to chronic cardiac inflammation. We found an increased presence of pro-inflammatory neutrophils and macrophages as well as neutrophils, macrophages and T-lymphocytes at 4-days and 8-weeks respectively post-MI in TgGRK5 hearts. Conversely, GRK5cKO mice were protected from ischemic injury and showed reduced early immune cell recruitment (predominantly monocytes) to the heart, improved contractility and reduced mortality compared to WT post-MI mice. Interestingly, cardiomyocyte-specific GRK2 transgenic mice did not share the same phenotype of TgGRK5 mice and did not have increased cardiac leukocyte migration and cytokine or chemokine production post-MI. CONCLUSIONS Our study shows that myocyte GRK5 has a crucial and GRK-selective role on the regulation of leucocyte infiltration into the heart, cardiac function and survival in a murine model of post-ischemic HF, supporting GRK5 inhibition as a therapeutic target for HF.
Collapse
Affiliation(s)
- Claudio de Lucia
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Giulia Borghetti
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Michela Piedepalumbo
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Jessica Ibetti
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Anna Maria Lucchese
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Eric W Barr
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Rajika Roy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Ama Dedo Okyere
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Haley Christine Murphy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via S. Pansini, 5, Naples, Italy
- Laboratory of neurovegetative system pathophysiology, Istituti Clinici Scientifici ICS Maugeri, IRCCS Istituto Scientifico di Telese Terme, Benevento, Italy
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
7
|
Lemoine KA, Fassas JM, Ohannesian SH, Purcell NH. On the PHLPPside: Emerging roles of PHLPP phosphatases in the heart. Cell Signal 2021; 86:110097. [PMID: 34320369 PMCID: PMC8403656 DOI: 10.1016/j.cellsig.2021.110097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
PH domain leucine-rich repeat protein phosphatase (PHLPP) is a family of enzymes made up of two isoforms (PHLPP1 and PHLPP2), whose actions modulate intracellular activity via the dephosphorylation of specific serine/threonine (Ser/Thr) residues on proteins such as Akt. Recent data generated in our lab, supported by findings from others, implicates the divergent roles of PHLPP1 and PHLPP2 in maintaining cellular homeostasis since dysregulation of these enzymes has been linked to various pathological states including cardiovascular disease, diabetes, ischemia/reperfusion injury, musculoskeletal disease, and cancer. Therefore, development of therapies to modulate specific isoforms of PHLPP could prove to be therapeutically beneficial in several diseases especially those targeting the cardiovascular system. This review is intended to provide a comprehensive summary of current literature detailing the role of the PHLPP isoforms in the development and progression of heart disease.
Collapse
Affiliation(s)
- Kellie A Lemoine
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Julianna M Fassas
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Shirag H Ohannesian
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Nicole H Purcell
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA; Cardiovascular Molecular Signaling, Huntington Medical Research Institutes, Pasadena, CA 91105, USA.
| |
Collapse
|
8
|
Metal dependent protein phosphatase PPM family in cardiac health and diseases. Cell Signal 2021; 85:110061. [PMID: 34091011 PMCID: PMC9107372 DOI: 10.1016/j.cellsig.2021.110061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/20/2022]
Abstract
Protein phosphorylation and dephosphorylation is central to signal transduction in nearly every aspect of cellular function, including cardiovascular regulation and diseases. While protein kinases are often regarded as the molecular drivers in cellular signaling with high specificity and tight regulation, dephosphorylation mediated by protein phosphatases is also gaining increasing appreciation as an important part of the signal transduction network essential for the robustness, specificity and homeostasis of cell signaling. Metal dependent protein phosphatases (PPM, also known as protein phosphatases type 2C, PP2C) belong to a highly conserved family of protein phosphatases with unique biochemical and molecular features. Accumulating evidence also indicates important and specific functions of individual PPM isoform in signaling and cellular processes, including proliferation, senescence, apoptosis and metabolism. At the physiological level, abnormal PPM expression and activity have been implicated in major human diseases, including cancer, neurological and cardiovascular disorders. Finally, inhibitors for some of the PPM members have been developed as a potential therapeutic strategy for human diseases. In this review, we will focus on the background information about the biochemical and molecular features of major PPM family members, with emphasis on their demonstrated or potential roles in cardiac pathophysiology. The current challenge and potential directions for future investigations will also be highlighted.
Collapse
|
9
|
Lian R, Zhang G, Yan S, Sun L, Zhang G. Identification of Molecular Regulatory Features and Markers for Acute Type A Aortic Dissection. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6697848. [PMID: 33953793 PMCID: PMC8057891 DOI: 10.1155/2021/6697848] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/03/2021] [Accepted: 03/25/2021] [Indexed: 12/03/2022]
Abstract
BACKGROUND Acute type A aortic dissection (ATAAD) is one of the most lethal cardiovascular diseases, and its molecular mechanism remains unclear. METHODS Differentially expressed genes (DEGs) between ATAAD and control were detected by limma R package in GSE52093, GSE153434, GSE98770, and GSE84827, respectively. The coexpression network of DEGs was identified by the WGCNA package. Enrichment analysis was performed for module genes that were positively correlated with ATAAD using clusterProfiler R package. In addition, differentially methylated markers between aortic dissection and control were identified by ChAMP package. After comparing with ATAAD-related genes, a protein-protein interaction (PPI) network was established based on the STRING database. The genes with the highest connectivity were identified as hub genes. Finally, differential immune cell infiltration between ATAAD and control was identified by ssGSEA. RESULTS From GSE52093 and GSE153434, 268 module genes were obtained with consistent direction of differential expression and high correlation with ATAAD. They were significantly enriched in T cell activation, HIF-1 signaling pathway, and cell cycle. In addition, 2060 differentially methylated markers were obtained from GSE84827. Among them, 77 methylation markers were ATAAD-related DEGs. Using the PPI network, we identified MYC, ITGA2, RND3, BCL2, and PHLPP2 as hub genes. Finally, we identified significantly differentially infiltrated immune cells in ATAAD. CONCLUSION The hub genes we identified may be regulated by methylation and participate in the development of ATAAD through immune inflammation and oxidative stress response. The findings may provide new insights into the molecular mechanisms and therapeutic targets for ATAAD.
Collapse
Affiliation(s)
- Rui Lian
- Graduate School of Peking Union Medical College, Beijing, China
- Emergency Department, China-Japan Friendship Hospital, Beijing, China
| | - Guochao Zhang
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Shengtao Yan
- Emergency Department, China-Japan Friendship Hospital, Beijing, China
| | - Lichao Sun
- Emergency Department, China-Japan Friendship Hospital, Beijing, China
| | - Guoqiang Zhang
- Graduate School of Peking Union Medical College, Beijing, China
- Emergency Department, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
10
|
Targeting GRK5 for Treating Chronic Degenerative Diseases. Int J Mol Sci 2021; 22:ijms22041920. [PMID: 33671974 PMCID: PMC7919044 DOI: 10.3390/ijms22041920] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/27/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell-surface receptors and they are responsible for the transduction of extracellular signals, regulating almost all aspects of mammalian physiology. These receptors are specifically regulated by a family of serine/threonine kinases, called GPCR kinases (GRKs). Given the biological role of GPCRs, it is not surprising that GRKs are also involved in several pathophysiological processes. Particular importance is emerging for GRK5, which is a multifunctional protein, expressed in different cell types, and it has been found located in single or multiple subcellular compartments. For instance, when anchored to the plasma membrane, GRK5 exerts its canonical function, regulating GPCRs. However, under certain conditions (e.g., pro-hypertrophic stimuli), GRK5 translocates to the nucleus of cells where it can interact with non-GPCR-related proteins as well as DNA itself to promote “non-canonical” signaling, including gene transcription. Importantly, due to these actions, several studies have demonstrated that GRK5 has a pivotal role in the pathogenesis of chronic-degenerative disorders. This is true in the cardiac cells, tumor cells, and neurons. For this reason, in this review article, we will inform the readers of the most recent evidence that supports the importance of targeting GRK5 to prevent the development or progression of cancer, cardiovascular, and neurological diseases.
Collapse
|
11
|
Fang F, Cheng L, Wu X, Ye M, Zhang H. miR-141 Promotes Colon Cancer Cell Proliferation by Targeted PHLPP2 Expression Inhibitionn. Cancer Manag Res 2020; 12:11341-11350. [PMID: 33204152 PMCID: PMC7661787 DOI: 10.2147/cmar.s256670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022] Open
Abstract
Objective Colon cancer (CC) is the third most common cancer with a high rate of incidence and mortality. Therefore, it is highly necessary to explore novel targets of CC. Methods The miRNA-seq and RNA-seq data of CC were accessed from the TCGA database. Differential analysis was performed using the "edgeR" package to identify differentially expressed miRNAs (DE_miRNAs). The downstream target genes of the target miRNA were then predicted by miRNA target prediction databases to identify the target mRNA. Normal colon cell line CCD-18Co and CC cell lines HCT-116, HT-29, SW620 and SW480 were chosen, and qRT-PCR was conducted to detect miR-141 expression in these cell lines. qRT-PCR and Western blot were carried out to determine PHLPP2 mRNA and protein expression, respectively. Dual-luciferase reporter gene assay was performed to verify the targeting relationship between miR-141 and PHLPP2 3'UTR. CCK-8 assay and colony formation assay were carried out to detect cell proliferation. Meanwhile, tumor xenograft model in nude mice was constructed to assess CC cell tumorigenic ability in vivo. Results miR-141 was markedly up-regulated in CC tissue. CC cell proliferation and in vivo tumorigenic ability were suppressed by miR-141 silencing but promoted by miR-141 over-expression. PHLPP2 was significantly down-regulated in cancer tissue. Dual-luciferase reporter gene assay indicated that miR-141 could bind to PHLPP2 3'UTR. PHLPP2 expression was noticeably elevated upon miR-141 deficiency but significantly inhibited upon miR-141 over-expression. CCK-8 and colony formation assay suggested that miR-141 facilitated CC cell proliferation by silencing PHLPP2. Conclusion miR-141 promotes CC cell proliferation by targeted silencing PHLPP2.
Collapse
Affiliation(s)
- Fazhuang Fang
- Department of Hepatobiliary, Pancreatic and Gastric Surgery, Jinhua Guangfu Hospital, Jinhua 321000, People's Republic of China
| | - Ling Cheng
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, People's Republic of China
| | - Xiaotang Wu
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, People's Republic of China
| | - Minfeng Ye
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, People's Republic of China
| | - Huizhong Zhang
- Department of Hepatobiliary, Pancreatic and Gastric Surgery, Jinhua Guangfu Hospital, Jinhua 321000, People's Republic of China
| |
Collapse
|
12
|
Wang H, Gu R, Tian F, Liu Y, Fan W, Xue G, Cai L, Xing Y. PHLPP2 as a novel metastatic and prognostic biomarker in non-small cell lung cancer patients. Thorac Cancer 2019; 10:2124-2132. [PMID: 31571378 PMCID: PMC6825916 DOI: 10.1111/1759-7714.13196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 12/24/2022] Open
Abstract
Background PH domain and leucine‐rich repeat protein phosphatase 2 (PHLPP2) has been reported to be a potent tumor suppressor in many human cancers. However, PHLPP2 has not been fully researched as a putative clinical prognostic biomarker of lung cancer. Methods The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases including data on 1383 non‐small cell lung cancer (NSCLC) patients were used to determine PHLPP2 expression. PHLPP2 expression was then examined by immunohistochemistry, and its clinical significance analyzed in 134 NSCLC patients, including 73 patients with adenocarcinoma and 81 with squamous cell carcinoma. Results We found PHLPP2 expression to be less pronounced in NSCLC tissue samples than that in nontumoral lung tissues according to data taken from TCGA and GEO datasets; this outcome was further validated by immunohistochemistry assay. The low PHLPP2 expression level was found to be associated with the presence of lymph node metastasis (P = 0.003). Importantly, PHLPP2 was found to be an independent indicator of prognosis for overall (hazard ratio [HR] = 0.520, 95% confidence interval [Cl] = 0.327–0.827; P = 0.006) and disease‐free survival (HR = 0.489, 95% Cl = 0.308–0.775; P = 0.002) in patients with surgically‐resected NSCLC by multivariate analysis. Conclusion Taken together, our findings show that PHLPP2 is a robust clinical marker for NSCLC survival and could serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Hongmei Wang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ruixue Gu
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fanglin Tian
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuechao Liu
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Weina Fan
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guiqin Xue
- General Surgical Department, The Fifth Hospital of Daqing, Daqing, China
| | - Li Cai
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ying Xing
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|