51
|
Protection of 6-OHDA neurotoxicity by PGF 2α through FP-ERK-Nrf2 signaling in SH-SY5Y cells. Toxicology 2021; 450:152686. [PMID: 33486071 DOI: 10.1016/j.tox.2021.152686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/26/2022]
Abstract
6-Hydroxydopamine (6-OHDA) is a neurotoxin that destroy dopaminergic neurons and widely used to establish animal models of Parkinson's disease. Prostaglandins (PGs) are involved in various cellular processes, including the damage and repair of neuronal cells. However, the function of PGF2α in neuronal cells remains unclear. In this study, we investigated the effects of PGF2α against 6-OHDA-mediated toxicity in human neuroblastoma SH-SY5Y cells and elucidated its underlying molecular mechanism. When the cells were treated with 6-OHDA (50 μM) for 6 h, the expression levels of PGF2α synthetic enzymes; cyclooxygenase-2 and aldo-keto reductase 1C3 as PGF2α synthase were enhanced in an incubation-time-dependent manner. In addition, the production of PGF2α was increased in 6-OHDA-treated cells. Fluprostenol, a PGF2α receptor (FP) agonist (500 nM), suppressed 6-OHDA-induced cell death by decreasing the production of reactive oxygen species (ROS) and increasing the expression of the anti-oxidant genes. These fluprostenol-mediated effects were inhibited by co-treatment with AL8810, an FP receptor antagonist (1 μM) or transfection with FP siRNA (20 nM). Moreover, 6-OHDA-induced phosphorylation of extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase family, was inhibited by co-incubation with AL8810. Furthermore, fluprostenol itself enhanced ERK phosphorylation and further elevated the 6-OHDA-induced phosphorylation of ERK. In addition, 6-OHDA induced nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), activating anti-oxidant gene expression, was repressed by co-culturing with AL8810. These results indicate that PGF2α suppressed 6-OHDA-induced neuronal cell death by enhancing anti-oxidant gene expression via the FP receptor-ERK-Nrf2 signaling. Thus, FP receptor is a potential target for inhibition of ROS-mediated neuronal cell death.
Collapse
|
52
|
Jaenen V, Fraguas S, Bijnens K, Heleven M, Artois T, Romero R, Smeets K, Cebrià F. Reactive oxygen species rescue regeneration after silencing the MAPK-ERK signaling pathway in Schmidtea mediterranea. Sci Rep 2021; 11:881. [PMID: 33441641 PMCID: PMC7806912 DOI: 10.1038/s41598-020-79588-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
Despite extensive research on molecular pathways controlling the process of regeneration in model organisms, little is known about the actual initiation signals necessary to induce regeneration. Recently, the activation of ERK signaling has been shown to be required to initiate regeneration in planarians. However, how ERK signaling is activated remains unknown. Reactive Oxygen Species (ROS) are well-known early signals necessary for regeneration in several models, including planarians. Still, the probable interplay between ROS and MAPK/ERK has not yet been described. Here, by interfering with major mediators (ROS, EGFR and MAPK/ERK), we were able to identify wound-induced ROS, and specifically H2O2, as upstream cues in the activation of regeneration. Our data demonstrate new relationships between regeneration-related ROS production and MAPK/ERK activation at the earliest regeneration stages, as well as the involvement of the EGFR-signaling pathway. Our results suggest that (1) ROS and/or H2O2 have the potential to rescue regeneration after MEK-inhibition, either by H2O2-treatment or light therapy, (2) ROS and/or H2O2 are required for the activation of MAPK/ERK signaling pathway, (3) the EGFR pathway can mediate ROS production and the activation of MAPK/ERK during planarian regeneration.
Collapse
Affiliation(s)
- V Jaenen
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - S Fraguas
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.,Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - K Bijnens
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - M Heleven
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - T Artois
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - R Romero
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain
| | - K Smeets
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium. .,Department of Biology and Geology, Faculty of Sciences, Agoralaan Building D, 3590, Diepenbeek, Belgium.
| | - F Cebrià
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain. .,Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
53
|
Tauffenberger A, Magistretti PJ. Reactive Oxygen Species: Beyond Their Reactive Behavior. Neurochem Res 2021; 46:77-87. [PMID: 33439432 PMCID: PMC7829243 DOI: 10.1007/s11064-020-03208-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/02/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022]
Abstract
Cellular homeostasis plays a critical role in how an organism will develop and age. Disruption of this fragile equilibrium is often associated with health degradation and ultimately, death. Reactive oxygen species (ROS) have been closely associated with health decline and neurological disorders, such as Alzheimer's disease or Parkinson's disease. ROS were first identified as by-products of the cellular activity, mainly mitochondrial respiration, and their high reactivity is linked to a disruption of macromolecules such as proteins, lipids and DNA. More recent research suggests more complex function of ROS, reaching far beyond the cellular dysfunction. ROS are active actors in most of the signaling cascades involved in cell development, proliferation and survival, constituting important second messengers. In the brain, their impact on neurons and astrocytes has been associated with synaptic plasticity and neuron survival. This review provides an overview of ROS function in cell signaling in the context of aging and degeneration in the brain and guarding the fragile balance between health and disease.
Collapse
Affiliation(s)
- Arnaud Tauffenberger
- King Abdullah University of Science and Technology, Thuwal, 23955, Kingdom of Saudi Arabia.
| | - Pierre J Magistretti
- King Abdullah University of Science and Technology, Thuwal, 23955, Kingdom of Saudi Arabia.
| |
Collapse
|
54
|
Chang CC, Liu YC, Lin CH, Liao YC. Histone acetyltransferase p300 mediates the upregulation of CTEN induced by the activation of EGFR signaling in cancer cells. Biochem Biophys Res Commun 2020; 534:53-58. [PMID: 33310188 DOI: 10.1016/j.bbrc.2020.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 01/08/2023]
Abstract
Upregulation of C-terminal tensin-like (CTEN) is induced by the activation of epidermal growth factor receptor (EGFR) signaling and mainly contributes to cancer cell migration and invasion. CTEN is known as a downstream target of the EGFR-RAF-MEK-ERK pathway but the regulatory mechanism underlying EGFR signaling regulates the increased expression of CTEN is still incompletely understood. In this study, we investigated the epigenetic regulation of CTEN gene transcription upon EGFR activation. Analyses of chromatin accessibility revealed that the structure of CTEN promoter became more loosed and the acetylation state of the histone tails within the core promoter region was increased after EGF treatment. Moreover, activation of EGFR signaling facilitates histone acetyltransferase p300 to be recruited to CTEN promoter through MEK-ERK pathway. MEK-ERK activation also induces the phosphorylation of p300, thereby enhancing the levels of histone acetylation within CTEN promoter, which in turn upregulates CTEN gene expression. Our work provides new insights into the actions of EGFR signaling to upregulate CTEN, which may lead to the rational design of novel therapeutic approaches.
Collapse
Affiliation(s)
- Chia-Chun Chang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Chou Liu
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Chih-Hsuan Lin
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Chun Liao
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
55
|
Nguyen D, Lin LY, Zhou JO, Kibby E, Sia TW, Tillis TD, Vapuryan N, Xu MR, Potluri R, Shin Y, Erler EA, Bronkema N, Boehlmer DJ, Chung CD, Burkhard C, Zeng SH, Grasso M, Acevedo LA, Marmorstein R, Fera D. Identification and Characterization of a B-Raf Kinase α-Helix Critical for the Activity of MEK Kinase in MAPK Signaling. Biochemistry 2020; 59:4755-4765. [PMID: 33272017 DOI: 10.1021/acs.biochem.0c00598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In the MAPK pathway, an oncogenic V600E mutation in B-Raf kinase causes the enzyme to be constitutively active, leading to aberrantly high phosphorylation levels of its downstream effectors, MEK and ERK kinases. The V600E mutation in B-Raf accounts for more than half of all melanomas and ∼3% of all cancers, and many drugs target the ATP binding site of the enzyme for its inhibition. Because B-Raf can develop resistance against these drugs and such drugs can induce paradoxical activation, drugs that target allosteric sites are needed. To identify other potential drug targets, we generated and kinetically characterized an active form of B-RafV600E expressed using a bacterial expression system. In doing so, we identified an α-helix on B-Raf, found at the B-Raf-MEK interface, that is critical for their interaction and the oncogenic activity of B-RafV600E. We assessed the binding between B-Raf mutants and MEK using pull downs and biolayer interferometry and assessed phosphorylation levels of MEK in vitro and in cells as well as its downstream target ERK to show that mutating certain residues on this α-helix is detrimental to binding and downstream activity. Our results suggest that this B-Raf α-helix binding site on MEK could be a site to target for drug development to treat B-RafV600E-induced melanomas.
Collapse
Affiliation(s)
- Diep Nguyen
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Linda Yingqi Lin
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Jeffrey O Zhou
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Emily Kibby
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Twan W Sia
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Tiara D Tillis
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Narine Vapuryan
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Ming-Ray Xu
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Rajiv Potluri
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - YongJoon Shin
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Elizabeth A Erler
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Naomi Bronkema
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Daniel J Boehlmer
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Christopher D Chung
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Caroline Burkhard
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| | - Shirley H Zeng
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael Grasso
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Lucila A Acevedo
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ronen Marmorstein
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Daniela Fera
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, Pennsylvania 19081, United States
| |
Collapse
|
56
|
Jin LQ, John BH, Hu J, Selzer ME. Activated Erk Is an Early Retrograde Signal After Spinal Cord Injury in the Lamprey. Front Neurosci 2020; 14:580692. [PMID: 33250705 PMCID: PMC7674770 DOI: 10.3389/fnins.2020.580692] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
We previously reported that spinal cord transection (TX) in the lamprey causes mRNA to accumulate in the injured tips of large reticulospinal (RS) axons. We sought to determine whether this mRNA accumulation results from phosphorylation and transport of retrograde signals, similar to what has been reported in mammalian peripheral nerve. Extracellular signal-regulated protein kinase (Erk), mediates the neurite outgrowth-promoting effects of many neurotrophic factors. To assess the role of Erk in retrograde signaling of RS axon injury, we used immunoblot and immunohistochemistry to determine the changes in phosphorylated Erk (p-Erk) in the spinal cord after spinal cord TX. Immunostaining for p-Erk increased within axons and local cell bodies, most heavily within the 1-2 mm closest to the TX site, at between 3 and 6 h post-TX. In axons, p-Erk was concentrated in 3-5 μm granules that became less numerous with distance from the TX. The retrograde molecular motor dynein colocalized with p-Erk, but vimentin, which in peripheral nerve was reported to participate with p-Erk as part of a retrograde signal complex, did not colocalize with p-Erk, even though vimentin levels were elevated post-TX. The results suggest that p-Erk, but not vimentin, may function as a retrograde axotomy signal in lamprey central nervous system neurons, and that this signal may induce transcription of mRNA, which is then transported down the axon to its injured tip.
Collapse
Affiliation(s)
- Li-Qing Jin
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Brittany H. John
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Jianli Hu
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Michael E. Selzer
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
57
|
Adams KL, Dahl KD, Gallo V, Macklin WB. Intrinsic and extrinsic regulators of oligodendrocyte progenitor proliferation and differentiation. Semin Cell Dev Biol 2020; 116:16-24. [PMID: 34110985 DOI: 10.1016/j.semcdb.2020.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022]
Abstract
Oligodendrocytes are highly specialized glial cells, responsible for producing myelin in the central nervous system (CNS). The multi-stage process of oligodendrocyte development is tightly regulated to ensure proper lineage progression of oligodendrocyte progenitor cells (OPCs) to mature myelin producing oligodendrocytes. This developmental process involves complex interactions between several intrinsic signaling pathways that are modulated by an array of extrinsic factors. Understanding these regulatory processes is of crucial importance, as it may help to identify specific molecular targets both to enhance plasticity in the normal CNS and to promote endogenous recovery following injury or disease. This review describes two major regulators that play important functional roles in distinct phases of oligodendrocyte development: OPC proliferation and differentiation. Specifically, we highlight the roles of the extracellular astrocyte/radial glia-derived protein Endothelin-1 in OPC proliferation and the intracellular Akt/mTOR pathway in OPC differentiation. Lastly, we reflect on how recent advances in neuroscience and scientific technology will enable greater understanding into how intrinsic and extrinsic regulators interact to generate oligodendrocyte diversity.
Collapse
Affiliation(s)
- Katrina L Adams
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Kristin D Dahl
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
58
|
Chen Y, Liu F, Chen BD, Li XM, Huang Y, Yu ZX, Gao XL, He CH, Yang YN, Ma YT, Gao XM. rAAV9-Mediated MEK1 Gene Expression Restores Post-conditioning Protection Against Ischemia Injury in Hypertrophic Myocardium. Cardiovasc Drugs Ther 2020; 34:3-14. [PMID: 32103377 DOI: 10.1007/s10557-020-06936-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE We investigated whether increased expression of activated mitogen-activated protein kinase (MAPK) kinases 1 (MEK1) restores ischemic post-conditioning (IPostC) protection in hypertrophic myocardium following ischemia/reperfusion (I/R) injury. METHODS C57Bl/6 mice received recombinant adeno-associated virus type 9 (rAAV9)-mediated activated MEK1 gene delivery systemically, then following the induction of cardiac hypertrophy via transverse aortic constriction for 4 weeks. In a Langendorff model, hypertrophic hearts were subjected to 40 min/60 min I/R or with IPostC intervention consisting of 6 cycles of 10 s reperfusion and 10 s no-flow before a 60-min reperfusion. Hemodynamics, infarct size (IS), myocyte apoptosis and changes in expression of reperfusion injury salvage kinase (RISK) pathway were examined. RESULTS rAAV9-MEK1 gene delivery led to a 4.3-fold and 2.7-fold increase in MEK1 mRNA and protein expression in the heart versus their control values. I/R resulted in a larger IS in hypertrophic than in non-hypertrophic hearts (52.3 ± 4.7% vs. 40.0 ± 2.5%, P < 0.05). IPostC mediated IS reduction in non-hypertrophic hearts (27.6 ± 2.6%, P < 0.05), while it had no significant effect in hypertrophic hearts (46.5 ± 3.1%, P=NS) compared with the IS in non-hypertrophic or hypertrophic hearts subjected to I/R injury only, respectively. Hemodynamic decline induced by I/R was preserved by IPostC in non-hypertrophic hearts but not in hypertrophic hearts. rAAV9-MEK1 gene delivery restored IPostC protection in hypertrophic hearts evidenced by reduced IS (32.0 ± 2.8% vs. 46.5 ± 3.1%) and cardiac cell apoptosis and largely preserved hemodynamic parameters. These protective effects were associated with significantly increased phosphorylation of ERK1/2 and ribosomal protein S6 kinases (p70S6K), but it had no influence on Akt and glycogen synthase kinase-3β. CONCLUSION These results demonstrated that rAAV9-mediated activated MEK1 expression restores IPostC protection in the hypertrophic heart against I/R injury through the activation of ERK pathway.
Collapse
Affiliation(s)
- You Chen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China
| | - Fen Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China.,Clinical Medical Research Institute, Xinjiang Medical University, Urumqi, 830054, China
| | - Bang-Dang Chen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China.,Clinical Medical Research Institute, Xinjiang Medical University, Urumqi, 830054, China
| | - Xiao-Mei Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China
| | - Ying Huang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China
| | - Zi-Xiang Yu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China
| | - Xiao-Li Gao
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Chun-Hui He
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China
| | - Yi-Ning Yang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China. .,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China.
| | - Yi-Tong Ma
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China. .,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China.
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China. .,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, China. .,Clinical Medical Research Institute, Xinjiang Medical University, Urumqi, 830054, China. .,Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, 830054, China.
| |
Collapse
|
59
|
Urushihara M, Kondo S, Kinoshita Y, Ozaki N, Jamba A, Nagai T, Fujioka K, Hattori T, Kagami S. (Pro)renin receptor promotes crescent formation via the ERK1/2 and Wnt/β-catenin pathways in glomerulonephritis. Am J Physiol Renal Physiol 2020; 319:F571-F578. [PMID: 32830537 DOI: 10.1152/ajprenal.00250.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
(Pro)renin receptor [(P)RR] has multiple functions, but its regulation and role in the pathogenesis in glomerulonephritis (GN) are poorly defined. The aims of the present study were to determine the effects of direct renin inhibition (DRI) and demonstrate the role of (P)RR on the progression of crescentic GN. The anti-glomerular basement membrane nephritis rat model developed progressive proteinuria (83.64 ± 10.49 mg/day) and glomerular crescent formation (percent glomerular crescent: 62.1 ± 2.3%) accompanied by increased macrophage infiltration and glomerular expression of monocyte chemoattractant protein (MCP)-1, (P)RR, phospho-extracellular signal-regulated kinase (ERK)1/2, Wnt4, and active β-catenin. Treatment with DRI ameliorated proteinuria (20.33 ± 5.88 mg/day) and markedly reduced glomerular crescent formation (20.9 ± 2.6%), induction of macrophage infiltration, (P)RR, phospho-ERK1/2, Wnt4, and active β-catenin. Furthermore, primary cultured parietal epithelial cells stimulated by recombinant prorenin showed significant increases in cell proliferation. Notably, while the ERK1/2 inhibitor PD98059 or (P)RR-specific siRNA treatment abolished the elevation in cell proliferation, DRI treatment did not abrogate this elevation. Moreover, cultured mesangial cells showed an increase in prorenin-induced MCP-1 expression. Interestingly, (P)RR or Wnt4-specific siRNA treatment or the β-catenin antagonist XAV939 inhibited the elevation of MCP-1 expression, whereas DRI did not. These results suggest that (P)RR regulates glomerular crescent formation via the ERK1/2 signaling and Wnt/β-catenin pathways during the course of anti-glomerular basement membrane nephritis and that DRI mitigates the progression of crescentic GN through the reduction of (P)RR expression but not inhibition of prorenin binding to (P)RR.
Collapse
Affiliation(s)
- Maki Urushihara
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| | - Shuji Kondo
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| | - Yukiko Kinoshita
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| | - Natsuko Ozaki
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| | - Ariunbold Jamba
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| | - Takashi Nagai
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| | - Keisuke Fujioka
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| | - Tomoki Hattori
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| | - Shoji Kagami
- Department of Pediatrics, Institute of Health Biosciences, The Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
60
|
Regulation of ERK basal and pulsatile activity control proliferation and exit from the stem cell compartment in mammalian epidermis. Proc Natl Acad Sci U S A 2020; 117:17796-17807. [PMID: 32651268 PMCID: PMC7395546 DOI: 10.1073/pnas.2006965117] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Understanding how intracellular signaling cascades control cell fate is a key issue in stem cell biology. Here we show that exit from the stem cell compartment in mammalian epidermis is characterized by pulsatile ERK MAPK activity. Basal activity and pulses are differentially regulated by DUSP10 and DUSP6, two phosphatases that have been shown previously to regulate differentiation commitment in the epidermis. ERK activity is controlled both transcriptionally and posttranscriptionally. Spatial segregation of mean ERK activity and pulses is observed both in reconstituted human epidermis and in mouse epidermis. Our findings demonstrate the tight spatial and temporal regulation of ERK MAPK expression and activity in mammalian epidermis. Fluctuation in signal transduction pathways is frequently observed during mammalian development. However, its role in regulating stem cells has not been explored. Here we tracked spatiotemporal ERK MAPK dynamics in human epidermal stem cells. While stem cells and differentiated cells were distinguished by high and low stable basal ERK activity, respectively, we also found cells with pulsatile ERK activity. Transitions from Basalhi-Pulselo (stem) to Basalhi-Pulsehi, Basalmid-Pulsehi, and Basallo-Pulselo (differentiated) cells occurred in expanding keratinocyte colonies and in response to differentiation stimuli. Pharmacological inhibition of ERK induced differentiation only when cells were in the Basalmid-Pulsehi state. Basal ERK activity and pulses were differentially regulated by DUSP10 and DUSP6, leading us to speculate that DUSP6-mediated ERK pulse down-regulation promotes initiation of differentiation, whereas DUSP10-mediated down-regulation of mean ERK activity promotes and stabilizes postcommitment differentiation. Levels of MAPK1/MAPK3 transcripts correlated with DUSP6 and DUSP10 transcripts in individual cells, suggesting that ERK activity is negatively regulated by transcriptional and posttranslational mechanisms. When cells were cultured on a topography that mimics the epidermal−dermal interface, spatial segregation of mean ERK activity and pulses was observed. In vivo imaging of mouse epidermis revealed a patterned distribution of basal cells with pulsatile ERK activity, and down-regulation was linked to the onset of differentiation. Our findings demonstrate that ERK MAPK signal fluctuations link kinase activity to stem cell dynamics.
Collapse
|
61
|
Ji J, Wu L, Feng J, Mo W, Wu J, Yu Q, Li S, Zhang J, Dai W, Xu X, Mao Y, Xu S, Chen K, Li J, Guo C. Cafestol preconditioning attenuates apoptosis and autophagy during hepatic ischemia-reperfusion injury by inhibiting ERK/PPARγ pathway. Int Immunopharmacol 2020; 84:106529. [PMID: 32344356 DOI: 10.1016/j.intimp.2020.106529] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/01/2020] [Accepted: 04/18/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The study was aimed to explore the hepatocellular protective functions of cafestol during hepatic ischemia-reperfusion injury and the possible mechanisms. METHODS Ninety male Balb/c mice were randomly divided into seven groups, including normal control group, L-cafestol(20mg/kg) group, H-cafestol(40mg/kg) group, sham group, IR group, L-cafestol(20mg/kg) + IR group, H-cafestol(40mg/kg) + IR group. Serum liver enzymes (ALT, AST), inflammation mediators, proteins associated with apoptosis and autophagy, indicators linked with ERK/PPARγ pathway, and liver histopathology were measured using ELISA, qRT-PCR, immunohistochemical staining, and western blotting at 2, 8, and 24 hours after reperfusion. RESULTS Our findings confirmed that cafestol preconditioning groups could reduce the levels of ALT and AST, alleviate liver pathological damage, suppress the release of inflammation mediators, inhibit the production of pro-apoptosis protein including caspase-3, caspase-9 and Bax, decrease the expression of autophagy-linked protein including Beclin-1 and LC3, increase anti-apoptosis protein Bcl-2, and restrain the activation of ERK and PPARγ. CONCLUSION Cafestol preconditioning could attenuate inflammatory response, apoptosis and autophagy on hepatic ischemia reperfusion injury by suppressing ERK/PPARγ pathway.
Collapse
Affiliation(s)
- Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Wenhui Mo
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jie Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China; Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai 200032, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China; Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China
| | - Xuanfu Xu
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, China
| | - Yuqing Mao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Shizan Xu
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Jinshan, Shanghai 201508, China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
62
|
Park YJ, Rahman MS, Pang WK, Ryu DY, Kim B, Pang MG. Bisphenol A affects the maturation and fertilization competence of spermatozoa. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 196:110512. [PMID: 32244115 DOI: 10.1016/j.ecoenv.2020.110512] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 06/11/2023]
Abstract
Although there are numerous studies on bisphenol A (BPA) on the testis and spermatozoa, the effect of BPA on the physiological link between the testis and maturation of spermatozoa has not been studied. To provide an optimal environment (acidic pH) for sperm maturation in the epididymis, clear cells secrete protons and principal cells reabsorb bicarbonate and the secreted proton. Because of its crucial role in sperm maturation and fertility, functional changes in the epididymis following BPA exposure must be considered to fully understand the mechanisms of BPA on male fertility. Here, we identified the adverse effects of BPA exposure during puberty in male mice. CD-1 male mice were gavaged daily with vehicle (corn oil) and 50 mg BPA/kg-BW for 6 weeks. We determined the changes in epididymis, functional sperm parameters including motility, capacitation status, tyrosine phosphorylation, and fertility-related protein expression and in vitro and in vivo fertility rate following BPA exposure. Expression of vacuolar-type H + -ATPase is necessary for the secretion of protons by clear cells of the caput epididymis and was directly down-regulated following BPA exposure, while there were no changes in the other epithelial cell types in the epididymis. Also, pERK 1/2 signaling pathway was increased significantly in the caput epididymis following BPA exposure. Consequently, the luminal pH slightly increased, resulting in premature capacitation of spermatozoa. Moreover, there was a significant loss of the acrosomal membrane following an increase of protein tyrosine phosphorylation, while PKA activity decreased during sperm capacitation. Fertility-related proteins also showed aberrant expression upon BPA exposure. These modifications resulted in decreased male fertility in vitro and in vivo.
Collapse
Affiliation(s)
- Yoo-Jin Park
- Department of Animal Science & Technology and BET Research Institute, Chgroung-Ang University, Anseong, Gyeonggi-do, 17546, Republic of Korea
| | - Md Saidur Rahman
- Department of Animal Science & Technology and BET Research Institute, Chgroung-Ang University, Anseong, Gyeonggi-do, 17546, Republic of Korea
| | - Won-Ki Pang
- Department of Animal Science & Technology and BET Research Institute, Chgroung-Ang University, Anseong, Gyeonggi-do, 17546, Republic of Korea
| | - Do-Yeal Ryu
- Department of Animal Science & Technology and BET Research Institute, Chgroung-Ang University, Anseong, Gyeonggi-do, 17546, Republic of Korea
| | - Bongki Kim
- Department of Animal Resources Science, Kongju National University, Yesan, Chungnam-do, 32439, Republic of Korea
| | - Myung-Geol Pang
- Department of Animal Science & Technology and BET Research Institute, Chgroung-Ang University, Anseong, Gyeonggi-do, 17546, Republic of Korea.
| |
Collapse
|
63
|
Lu Y, Liu B, Liu Y, Yu X, Cheng G. Dual effects of active ERK in cancer: A potential target for enhancing radiosensitivity. Oncol Lett 2020; 20:993-1000. [PMID: 32724338 PMCID: PMC7377092 DOI: 10.3892/ol.2020.11684] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/20/2020] [Indexed: 12/20/2022] Open
Abstract
Ionizing radiation (IR) is an important cancer treatment approach. However, radioresistance eventually occurs, resulting in poor outcomes in patients with cancer. Radioresistance is associated with multiple signaling pathways, particularly pro-survival signaling pathways. The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade is an important signaling pathway that initiates several cellular processes and is regulated by various stimuli, including IR. Although numerous studies have demonstrated the pro-survival effects of active ERK, activation of ERK has also been associated with cell death, indicating that radiosensitization may occur by ERK stimulation. In this context, the present review describes the associations between ERK signaling, cancer and IR, and discusses the association between ERK and its pro-survival function in cancer cells, including stimuli, molecular mechanisms, clinical use of inhibitors and underlying limitations. Additionally, the present review introduces the view that active ERK may induce cell death, and describes the potential factors associated with this process. This review describes the various outcomes induced by active ERK to prompt future studies to aim to enhance radiosensitivity in the treatment of cancer.
Collapse
Affiliation(s)
- Yinliang Lu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Baocai Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ying Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xinyue Yu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Guanghui Cheng
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
64
|
Peng W, Mo X, Li L, Lu T, Hu Z. PAQR3 protects against oxygen-glucose deprivation/reperfusion-induced injury through the ERK signaling pathway in N2A cells. J Mol Histol 2020; 51:307-315. [PMID: 32448978 DOI: 10.1007/s10735-020-09881-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
Cerebral ischemia-reperfusion injury is pivotal in the development of multiple-subcellular organelle and tissue injury after acute ischemic stroke. Recently, the Golgi apparatus (GA) has been shown to be a key subcellular organelle that plays an important role in neuroprotection against oxygen-glucose deprivation/reperfusion (OGD/R) injury. PAQR3, a scaffold protein exclusively localized in the GA, was originally discovered as a potential tumor suppressor protein. PAQR3 acts as a spatial regulator of Raf-1 that binds Raf-1 and sequesters it to the GA, where it negatively modulates the Ras/Raf/MEK/ERK signaling pathway in tumor models. Studies suggest that suppression of the ERK pathway can alleviate OGD/R-induced cell apoptosis. However, whether PAQR3 has potential effects on ischemic stroke and the underlying mechanism(s) remain unexplored. The current study is the first to show that PAQR3 was significantly downregulated in mouse neuroblastoma (N2A) cells upon OGD/R exposure, both at the mRNA and protein levels. Compared to that in controls, the mRNA level of PAQR3 began to decline at 0 h (0 h) after reperfusion, while the protein level began to decline at 4 h. Furthermore, overexpression of PAQR3 reduced OGD/R-induced apoptosis. The mRNA and protein levels of total ERK1 and ERK2 were unaltered, while activated p-ERK1 and p-ERK2 were decreased in N2A cells transfected with a PAQR3 expression vector after OGD for 4 h plus 24 h of reperfusion. Collectively, these data indicated that increased PAQR3 expression protected against OGD/R-induced apoptosis possibly by inhibiting the ERK signaling pathway. Therefore, PAQR3 might be a new attractive target in the treatment of OGD/R insult, and the underlying mechanism will pave the way for its potential experimental and clinical application.
Collapse
Affiliation(s)
- Wenna Peng
- Department of Rehabilitation, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoye Mo
- Department of Emergency, First Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lihua Li
- Colleges of Medicine, Jishou University, Jishou, Hunan, China
| | - Tonglin Lu
- Department of Intensive Care Unit, Hunan Provincial People's Hospital, Hunan Normal University, Changsha, Hunan, China
| | - Zhiping Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
65
|
Raha S, Kim SM, Lee HJ, Yumnam S, Saralamma VV, Ha SE, Lee WS, Kim GS. Naringin Induces Lysosomal Permeabilization and Autophagy Cell Death in AGS Gastric Cancer Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:679-702. [PMID: 32329644 DOI: 10.1142/s0192415x20500342] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Autophagy is a process of active programmed cell death, where a dying cell induces autophagosomes and subsequently regulated by degradative machinery. The aim of this study was to investigate the mechanism behind induction of autophagic cell death by Naringin flavonoid in AGS cancer cells. Growth inhibition of AGS cells showed downregulation of PI3K/Akt/mTOR signaling by Naringin treatment. Transmission electron microscopy observation showed swollen mitochondria and lysosome near peri-nuclear zone fused with autophagic vacuoles. Rapamycin pre-treatment with Naringin showed significant decrease in mTOR phosphorylation and increase in LC3B activation in AGS cells. Decrease in mTOR phosphorylation is associated with lysosomal function activation was observed by time-dependent treatment of Naringin. Induction of lysosomal membrane permeabilization (LMP) was observed by LAMP1 activation leading lysosomal cell death by releasing Cathepsin D from lysosomal lumen to cytosol. Naringin treated AGS cells showed up-regulating BH3 domain Bad, down-regulating Bcl-xL, and Bad phosphorylation and significant mitochondrial fluorescence intensity expression. Significant localization of mitochondria and LC3B activation was examined by person coefficient correlation. Activation of ERK1/2-p38 MAPKs and production of intracellular ROS has been observed over Naringin treatment. It has also been elucidated that pre-treatment with NAC inhibited mitochondria-LC3B colocalization, where ROS acted as upstream of ERK1/2-p38 MAPKs activation. Lysosomal cell death involvement has been evaluated by BAF A1 pre-treatment, inhibiting LAMP1, Cathepsin D, ROS, and blocking autophagolysosome in AGS cell death. Taken together, these findings show that, Naringin induced autophagy cell death involves LMP mediated lysosomal damage and BH3 protein Bad activation in AGS cancer cells.
Collapse
Affiliation(s)
- Suchismita Raha
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea.,Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University, School of Medicine, 90 Chilam-dong, Jinju 52727, Republic of Korea
| | - Seong Min Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Ho Jeong Lee
- Biological Resources Research Group, Bioenvironmental Science & Toxicology, Division, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju 52834, Republic of Korea
| | - Silvia Yumnam
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea.,College of Pharmacy, Gachon University, 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea
| | - Venu VenkatarameGowda Saralamma
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Sang Eun Ha
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Won Sup Lee
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea.,Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University, School of Medicine, 90 Chilam-dong, Jinju 52727, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| |
Collapse
|
66
|
Yuan Z, Wang Y, Yu W, Xie W, Zhang Z, Wang J, Zhang H, Han Y, Weng Q. Seasonal expressions of oxytocin and oxytocin receptor in the epididymides in the wild ground squirrels (Citellus Dauricus Brandt). Gen Comp Endocrinol 2020; 289:113391. [PMID: 31917151 DOI: 10.1016/j.ygcen.2020.113391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/02/2019] [Accepted: 01/03/2020] [Indexed: 12/19/2022]
Abstract
The aim of this study was to detect the seasonal expressions of oxytocin (OT), oxytocin receptor (OTR), extracellular signal-regulated kinase1 and 2 (ERK1/2) and phospho-ERK1/2 (pERK1/2) in the epididymis of the wild ground squirrels (Citellus Dauricus Brandt) during the breeding season and non-breeding season. Histological results showed that size, weight, cell number and lumen diameter of epididymis underwent acute seasonal changes, which were all peaked in the breeding season. Immunohistochemical results suggested that strong staining of OT, OTR, ERK1/2, and pERK1/2 were observed in the epithelial layer in the whole epididymis, along with intense OT and OTR signal in smooth muscle cell (Smc) in caudal epididymis in the breeding season. The protein expression levels of OTR, ERK1/2, and pERK1/2 in the epididymis were higher in the breeding season than those of the non-breeding season. Besides, hormone assay revealed that there was no significant serum concentration of OT in these two periods, while epididymal concentration showed higher value in the breeding season. In summary, the identified localization and local concentration of OT in the epididymis in the wild ground squirrel suggested that epididymis may assume as a source of OT, and OT could act via OTR to activate ERK1/2 signaling to regulate seasonal epididymal functions.
Collapse
Affiliation(s)
- Zhengrong Yuan
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Yuhan Wang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Wenyang Yu
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Wenqian Xie
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Ziwen Zhang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Junjie Wang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Haolin Zhang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Yingying Han
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Qiang Weng
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China.
| |
Collapse
|
67
|
Fathi E, Valipour B, Vietor I, Farahzadi R. An overview of the myocardial regeneration potential of cardiac c-Kit + progenitor cells via PI3K and MAPK signaling pathways. Future Cardiol 2020; 16:199-209. [PMID: 32125173 DOI: 10.2217/fca-2018-0049] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In recent years, several studies have investigated cell transplantation as an innovative strategy to restore cardiac function following heart failure. Previous studies have also shown cardiac progenitor cells as suitable candidates for cardiac cell therapy compared with other stem cells. Cellular kit (c-kit) plays an important role in the survival and migration of cardiac progenitor cells. Like other types of cells, in the heart, cellular responses to various stimuli are mediated via coordinated pathways. Activation of c-kit+ cells leads to subsequent activation of several downstream mediators such as PI3K and the MAPK pathways. This review aims to outline current research findings on the role of PI3K/AKT and the MAPK pathways in myocardial regeneration potential of c-kit+.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Behnaz Valipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ilja Vietor
- Division of Cell Biology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria
| | - Raheleh Farahzadi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran.,Hematology & Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
68
|
Current Advances in the Treatment of BRAF-Mutant Melanoma. Cancers (Basel) 2020; 12:cancers12020482. [PMID: 32092958 PMCID: PMC7072236 DOI: 10.3390/cancers12020482] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/02/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma is the most lethal form of skin cancer. Melanoma is usually curable with surgery if detected early, however, treatment options for patients with metastatic melanoma are limited and the five-year survival rate for metastatic melanoma had been 15-20% before the advent of immunotherapy. Treatment with immune checkpoint inhibitors has increased long-term survival outcomes in patients with advanced melanoma to as high as 50% although individual response can vary greatly. A mutation within the MAPK pathway leads to uncontrollable growth and ultimately develops into cancer. The most common driver mutation that leads to this characteristic overactivation in the MAPK pathway is the B-RAF mutation. Current combinations of BRAF and MEK inhibitors that have demonstrated improved patient outcomes include dabrafenib with trametinib, vemurafenib with cobimetinib or encorafenib with binimetinib. Treatment with BRAF and MEK inhibitors has met challenges as patient responses began to drop due to the development of resistance to these inhibitors which paved the way for development of immunotherapies and other small molecule inhibitor approaches to address this. Resistance to these inhibitors continues to push the need to expand our understanding of novel mechanisms of resistance associated with treatment therapies. This review focuses on the current landscape of how resistance occurs with the chronic use of BRAF and MEK inhibitors in BRAF-mutant melanoma and progress made in the fields of immunotherapies and other small molecules when used alone or in combination with BRAF and MEK inhibitors to delay or circumvent the onset of resistance for patients with stage III/IV BRAF mutant melanoma.
Collapse
|
69
|
Meng X, Liu P, Wu Y, Liu X, Huang Y, Yu B, Han J, Jin H, Tan X. Integrin beta 4 (ITGB4) and its tyrosine-1510 phosphorylation promote pancreatic tumorigenesis and regulate the MEK1-ERK1/2 signaling pathway. Bosn J Basic Med Sci 2020; 20:106-116. [PMID: 31242404 PMCID: PMC7029197 DOI: 10.17305/bjbms.2019.4255] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer death, with a 5-year survival rate of only 1–4%. Integrin-mediated cell adhesion is critical for the initiation, progression, and metastasis of cancer. In this study we investigated the role of integrin β4 (ITGB4) and its phosphorylation at tyrosine Y1510 (p-ITGB4-Y1510) in the tumorigenesis of pancreatic cancer. We analyzed the expression of ITGB4 and p-ITGB4-Y1510 in pancreatic cancer tissue and cell lines using immunohistochemistry, Western blot, or semi-quantitative reverse transcription PCR. ITGB4 and p-ITGB4-Y1510 were highly expressed in pancreatic cancer (n = 176) compared with normal pancreatic tissue (n = 171). High p-ITGB4-Y1510 expression correlated with local invasion and distant metastasis of pancreatic cancer, and high ITGB4 was significantly associated with poor survival of patients. Inhibition of ITGB4 by siRNA significantly reduced migration and invasion of PC-1.0 and AsPC-1 cells. Overexpression of the mutant ITGB4-Y1510A (a mutation of tyrosine to alanine at 1510 position) in PC-1.0 and AsPC-1 cells not only blocked the ITGB4 phosphorylation at Y1510 but also suppressed the expression of ITGB4 (p < 0.05 vs. wild-type ITGB4). The transfection of PC-1.0 and AsPC-1 cells with ITGB4-Y1510A significantly decreased the level of p-mitogen-activated protein kinase kinase (MEK)1 (T292) and p-extracellular signal-regulated kinase (ERK)1/2 but did not affect the level of p-MEK1 (T386) and p-MEK2 (T394). Overall, our study showed that ITGB4 and its phosphorylated form promote cell migration and invasion in pancreatic cancer and that p-ITGB4-Y1510 regulates the downstream MEK1-ERK1/2 signaling cascades. Targeting ITGB4 or its phosphorylation at Y1510 may be a novel therapeutic option for pancreatic cancer.
Collapse
Affiliation(s)
- Xiangli Meng
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Peng Liu
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Yunhao Wu
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Xinlu Liu
- Department of Anus and Intestine Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yinpeng Huang
- Minimally Invasive Area of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Boqiang Yu
- Department of General Surgery, Fushun Central Hospital, Fushun, China
| | - Jiahong Han
- Department of Surgery, Liaoning Electric Power Center Hospital, Shenyang, China.
| | - Haoyi Jin
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China.
| | - Xiaodong Tan
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China.
| |
Collapse
|
70
|
Das Ghosh L, Hasan J, Jain A, Sundaresan NR, Chatterjee K. A nanopillar array on black titanium prepared by reactive ion etching augments cardiomyogenic commitment of stem cells. NANOSCALE 2019; 11:20766-20776. [PMID: 31651003 DOI: 10.1039/c9nr03424b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A major impediment in the clinical translation of stem cell therapy has been the inability to efficiently and reproducibly direct differentiation of a large population of stem cells. Thus, we aimed to engineer a substrate for culturing stem cells to efficiently induce cardiomyogenic lineage commitment. In this work, we present a nanopillar array on the surface of titanium that was prepared by mask-less reactive ion etching. Scanning electron and atomic force microscopy revealed that the surface was covered by vertically aligned nanopillars each of ≈1 μm with a diameter of ≈80 nm. The nanopillars supported the attachment and proliferation of human mesenchymal stem cells (hMSCs). Cardiomyogenic lineage commitment of the stem cells was more enhanced on the nanopillars than on the smooth surface. When co-cultured with neonatal rat cardiomyocytes, the cyclic pattern of calcium transport observed distinctly in cells differentiated on the arrays compared to the cells cultured on the smooth surface was the functional validation of differentiation. The use of small molecule inhibitors revealed that integrins namely, α2β1 and αvβ3, are essential for cardiomyogenesis on the nanostructured surface, which is further mediated by FAK, Erk and Akt cell signaling pathways. This study demonstrates that the nanopillar array efficiently promotes the cardiomyogenic lineage commitment of stem cells via integrin-mediated signaling and can potentially serve as a platform for the ex vivo differentiation of stem cells toward cell therapy in cardiac tissue repair and regeneration.
Collapse
Affiliation(s)
- Lopamudra Das Ghosh
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - Jafar Hasan
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - Aditi Jain
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - Nagalingam R Sundaresan
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India. and Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India. and Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
71
|
The Effects of TGF-β Signaling on Cancer Cells and Cancer Stem Cells in the Bone Microenvironment. Int J Mol Sci 2019; 20:ijms20205117. [PMID: 31619018 PMCID: PMC6829436 DOI: 10.3390/ijms20205117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/02/2019] [Accepted: 10/14/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) plays a key role in bone metastasis formation; we hypothesized the possible involvement of TGF-β in the induction of cancer stem cells (CSCs) in the bone microenvironment (micro-E), which may be responsible for chemo-resistance. METHODS Mouse mammary tumor cells were implanted under the dorsal skin flap over the calvaria and into a subcutaneous (subQ) lesions in female mice, generating tumors in the bone and subQ micro-Es. After implantation of the tumor cells, mice were treated with a TGF-β R1 kinase inhibitor (R1-Ki). RESULTS Treatment with R1-Ki decreased tumor volume and cell proliferation in the bone micro-E, but not in the subQ micro-E. R1-Ki treatment did not affect the induction of necrosis or apoptosis in either bone or subQ micro-E. The number of cells positive for the CSC markers, SOX2, and CD166 in the bone micro-E, were significantly higher than those in the subQ micro-E. R1-Ki treatment significantly decreased the number of CSC marker positive cells in the bone micro-E but not in the subQ micro-E. TGF-β activation of the MAPK/ERK and AKT pathways was the underlying mechanism of cell proliferation in the bone micro-E. BMP signaling did not play a role in cell proliferation in either micro-E. CONCLUSION Our results indicated that the bone micro-E is a key niche for CSC generation, and TGF-β signaling has important roles in generating CSCs and tumor cell proliferation in the bone micro-E. Therefore, it is critically important to evaluate responses to chemotherapeutic agents on both cancer stem cells and proliferating tumor cells in different tumor microenvironments in vivo.
Collapse
|
72
|
Wang Q, Onuma K, Liu C, Wong H, Bloom MS, Elliott EE, Cao RR, Hu N, Lingampalli N, Sharpe O, Zhao X, Sohn DH, Lepus CM, Sokolove J, Mao R, Cisar CT, Raghu H, Chu CR, Giori NJ, Willingham SB, Prohaska SS, Cheng Z, Weissman IL, Robinson WH. Dysregulated integrin αVβ3 and CD47 signaling promotes joint inflammation, cartilage breakdown, and progression of osteoarthritis. JCI Insight 2019; 4:128616. [PMID: 31534047 PMCID: PMC6795293 DOI: 10.1172/jci.insight.128616] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is the leading cause of joint failure, yet the underlying mechanisms remain elusive, and no approved therapies that slow progression exist. Dysregulated integrin function was previously implicated in OA pathogenesis. However, the roles of integrin αVβ3 and the integrin-associated receptor CD47 in OA remain largely unknown. Here, transcriptomic and proteomic analyses of human and murine osteoarthritic tissues revealed dysregulated expression of αVβ3, CD47, and their ligands. Using genetically deficient mice and pharmacologic inhibitors, we showed that αVβ3, CD47, and the downstream signaling molecules Fyn and FAK are crucial to OA pathogenesis. MicroPET/CT imaging of a mouse model showed elevated ligand-binding capacities of integrin αVβ3 and CD47 in osteoarthritic joints. Further, our in vitro studies demonstrated that chondrocyte breakdown products, derived from articular cartilage of individuals with OA, induced αVβ3/CD47-dependent expression of inflammatory and degradative mediators, and revealed the downstream signaling network. Our findings identify a central role for dysregulated αVβ3 and CD47 signaling in OA pathogenesis and suggest that activation of αVβ3 and CD47 signaling in many articular cell types contributes to inflammation and joint destruction in OA. Thus, the data presented here provide a rationale for targeting αVβ3, CD47, and their signaling pathways as a disease-modifying therapy.
Collapse
Affiliation(s)
- Qian Wang
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Kazuhiro Onuma
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Changhao Liu
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, California, USA
| | - Heidi Wong
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Eileen E. Elliott
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Richard R.L. Cao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Nick Hu
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Nithya Lingampalli
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Orr Sharpe
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Xiaoyan Zhao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Dong Hyun Sohn
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do, South Korea
| | - Christin M. Lepus
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Jeremy Sokolove
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Rong Mao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Cecilia T. Cisar
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Harini Raghu
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Constance R. Chu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopedic Surgery
| | - Nicholas J. Giori
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopedic Surgery
| | - Stephen B. Willingham
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Susan S. Prohaska
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, California, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
73
|
Wei Y, Zhang X, Wen S, Huang S, Huang Q, Lu S, Bai F, Nie J, Wei J, Lu Z, Lin X. Methyl helicterate inhibits hepatic stellate cell activation through downregulating the ERK1/2 signaling pathway. J Cell Biochem 2019; 120:14936-14945. [PMID: 31009108 DOI: 10.1002/jcb.28756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/22/2018] [Accepted: 01/07/2019] [Indexed: 11/11/2022]
Abstract
The present study was to investigate the inhibitory effect of methyl helicterate (MH) on hepatic stellate cells (HSC-T6), primarily elucidating the underlying mechanism of MH against liver fibrosis. HSC-T6 cells were activated by platelet-derived growth factor (PDGF) stimulation, and then the effects of MH on cell viability, cytomembrane integrity, colony, migration, apoptosis, and cell cycle were detected. Moreover, the regulative mechanism of MH on HSCs was investigated by detecting the activation of the extracellular signal-regulated kinase (ERK1/2) signaling pathway. The results showed that MH significantly inhibited HSC-T6 cell viability and proliferation in a concentration-dependent manner. It notably promoted the release of lactate dehydrogenase, destroying cell membrane integrity. MH also markedly inhibited HSC-T6 cell clonogenicity and migration. Moreover, MH treatment significantly induced cell apoptosis and arrested cell cycle at the G2 phase. The further study showed that MH inhibited the expression of ERK1, ERK2, c-fos, c-myc, and Ets-1, blocking the ERK1/2 pathway. In conclusion, this study demonstrates that MH significantly inhibits HSC activation and promotes cell apoptosis via downregulation of the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Wei
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Xiaolin Zhang
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Shujuan Wen
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Shaode Huang
- Pharmaceutical College, Guangxi Agricultural Vocational College, Nanning, China
| | - Quanfang Huang
- Department of Pharmacy, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Shengjuan Lu
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Facheng Bai
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Jinlan Nie
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Jinbin Wei
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Zhongpeng Lu
- Department of Pharmacy, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
- Pharmaceutical College, University of Arkansas Medical School, Little Rock, Arkansas
| | - Xing Lin
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University, Nanning, China
| |
Collapse
|
74
|
Flynn JK, Dankers W, Morand EF. Could GILZ Be the Answer to Glucocorticoid Toxicity in Lupus? Front Immunol 2019; 10:1684. [PMID: 31379872 PMCID: PMC6652235 DOI: 10.3389/fimmu.2019.01684] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GC) are used globally to treat autoimmune and inflammatory disorders. Their anti-inflammatory actions are mainly mediated via binding to the glucocorticoid receptor (GR), creating a GC/GR complex, which acts in both the cytoplasm and nucleus to regulate the transcription of a host of target genes. As a result, signaling pathways such as NF-κB and AP-1 are inhibited, and cell activation, differentiation and survival and cytokine and chemokine production are suppressed. However, the gene regulation by GC can also cause severe side effects in patients. Systemic lupus erythematosus (SLE or lupus) is a multisystem autoimmune disease, characterized by a poorly regulated immune response leading to chronic inflammation and dysfunction of multiple organs, for which GC is the major current therapy. Long-term GC use, however, can cause debilitating adverse consequences for patients including diabetes, cardiovascular disease and osteoporosis and contributes to irreversible organ damage. To date, there is no alternative treatment which can replicate the rapid effects of GC across multiple immune cell functions, effecting disease control during disease flares. Research efforts have focused on finding alternatives to GC, which display similar immunoregulatory actions, without the devastating adverse metabolic effects. One potential candidate is the glucocorticoid-induced leucine zipper (GILZ). GILZ is induced by low concentrations of GC and is shown to mimic the action of GC in several inflammatory processes, reducing immunity and inflammation in in vitro and in vivo studies. Additionally, GILZ has, similar to the GC-GR complex, the ability to bind to both NF-κB and AP-1 as well as DNA directly, to regulate immune cell function, while potentially lacking the GC-related side effects. Importantly, in SLE patients GILZ is under-expressed and correlates negatively with disease activity, suggesting an important regulatory role of GILZ in SLE. Here we provide an overview of the actions and use of GC in lupus, and discuss whether the regulatory mechanisms of GILZ could lead to the development of a novel therapeutic for lupus. Increased understanding of the mechanisms of action of GILZ, and its ability to regulate immune events leading to lupus disease activity has important clinical implications for the development of safer anti-inflammatory therapies.
Collapse
Affiliation(s)
- Jacqueline K Flynn
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Wendy Dankers
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Eric F Morand
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
75
|
Wang T, Wang CJ, Tian S, Song HB. Overexpressed IGFBP5 promotes cell proliferation and inhibits apoptosis of nucleus pulposus derived from rats with disc degeneration through inactivating the ERK/MAPK axis. J Cell Biochem 2019; 120:18782-18792. [PMID: 31310371 DOI: 10.1002/jcb.29191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/29/2019] [Indexed: 12/19/2022]
Abstract
It is previously suggested that insulin-like growth factor binding proteins (IGFBPs) potentially share an association with disc degeneration (DD) that causes back pain. This study aimed at exploring the functional relevance of IGFBP5 in DD by establishing a rat model of DD. The nucleus pulposus (NP) cells were transduced with IGFBP5-shRNA or IGFBP5 overexpression to determine the cellular processes (proliferation, apoptosis, as well as colony formation). The protein levels of apoptosis-related proteins were evaluated. Furthermore, NP cells were treated with the extracellular signal-regulated kinases/mitogen-activated protein kinase (ERK/MAPK) pathway inhibitor (PD98059) followed by measurement of ERK protein level and ERK phosphorylation content. The NP cells showed suppressed proliferation and colony formation ability, yet promoted apoptosis after transfection with IGFBP5-shRNA. It was found that silencing of IGFBP5 could lead to the ERK/MAPK axis activation, as indicated by an elevated ERK protein level and ERK phosphorylation content. However, overexpression of IGFBP5 could reverse all the reaction induced by silenced IGFBP5. These key findings demonstrate that overexpressed IGFBP5 inactivates the ERK/MAPK axis to stimulate the proliferation and inhibit apoptosis of NP cells in a rat model of DD.
Collapse
Affiliation(s)
- Tao Wang
- Department of Spine Surgery, Dongying People's Hospital, Dongying, Shandong, P.R. China
| | - Chun-Ju Wang
- Department of Spine Surgery, Dongying People's Hospital, Dongying, Shandong, P.R. China
| | - Shuang Tian
- Department of Spine Surgery, Dongying People's Hospital, Dongying, Shandong, P.R. China
| | - Hai-Bo Song
- Department of Spine Surgery, Dongying People's Hospital, Dongying, Shandong, P.R. China
| |
Collapse
|
76
|
Olea-Flores M, Zuñiga-Eulogio MD, Mendoza-Catalán MA, Rodríguez-Ruiz HA, Castañeda-Saucedo E, Ortuño-Pineda C, Padilla-Benavides T, Navarro-Tito N. Extracellular-Signal Regulated Kinase: A Central Molecule Driving Epithelial-Mesenchymal Transition in Cancer. Int J Mol Sci 2019; 20:E2885. [PMID: 31200510 PMCID: PMC6627365 DOI: 10.3390/ijms20122885] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 12/18/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a reversible cellular process, characterized by changes in gene expression and activation of proteins, favoring the trans-differentiation of the epithelial phenotype to a mesenchymal phenotype. This process increases cell migration and invasion of tumor cells, progression of the cell cycle, and resistance to apoptosis and chemotherapy, all of which support tumor progression. One of the signaling pathways involved in tumor progression is the MAPK pathway. Within this family, the ERK subfamily of proteins is known for its contributions to EMT. The ERK subfamily is divided into typical (ERK 1/2/5), and atypical (ERK 3/4/7/8) members. These kinases are overexpressed and hyperactive in various types of cancer. They regulate diverse cellular processes such as proliferation, migration, metastasis, resistance to chemotherapy, and EMT. In this context, in vitro and in vivo assays, as well as studies in human patients, have shown that ERK favors the expression, function, and subcellular relocalization of various proteins that regulate EMT, thus promoting tumor progression. In this review, we discuss the mechanistic roles of the ERK subfamily members in EMT and tumor progression in diverse biological systems.
Collapse
Affiliation(s)
- Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Miriam Daniela Zuñiga-Eulogio
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Miguel Angel Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Hugo Alberto Rodríguez-Ruiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Carlos Ortuño-Pineda
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| |
Collapse
|
77
|
Li W, He W, Xia P, Sun W, Shi M, Zhou Y, Zhu W, Zhang L, Liu B, Zhu J, Zhu Y, Zhou E, Sun M, Gao K. Total Extracts of Abelmoschus manihot L. Attenuates Adriamycin-Induced Renal Tubule Injury via Suppression of ROS-ERK1/2-Mediated NLRP3 Inflammasome Activation. Front Pharmacol 2019; 10:567. [PMID: 31191310 PMCID: PMC6548014 DOI: 10.3389/fphar.2019.00567] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/06/2019] [Indexed: 12/11/2022] Open
Abstract
Abelmoschus manihot (L.) Medik. (Malvaceae) is a herb used in traditional Chinese medicine to treat some kidney diseases. To date, the detailed mechanisms by which A. manihot improves some kinds of renal disease are not fully understood. In this study, we established Adriamycin-induced NRK-52E cells, the normal rat kidney epithelial cell line, injury, and Sprague-Dawley rats with Adriamycin-induced nephropathy to evaluate the role and mechanisms of total extracts of A. manihot flower (TEA) both in vitro and in vivo. We found that TEA ameliorated Adriamycin-induced cellular morphological changes, cell viability, and apoptosis through the suppression of protein oxidation and ERK1/2 signaling. However, this anti-oxidative stress role of TEA was independent of ROS inhibition. Adriamycin activated ERK1/2 signaling followed by activation of NLRP3 inflammasomes. TEA suppressed NLRP3 inflammasomes via inhibition of ERK1/2 signal transduction; decreased proteinuria and attenuated renal tubule lesions; and inhibited the expression of NLRP3 in tubules in rats with Adriamycin nephropathy. Collectively, TEA protects renal tubular cells against Adriamycin-induced tubule injury via inhibition of ROS-ERK1/2-NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Wei Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Weiming He
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Ping Xia
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Wei Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Ming Shi
- Division of Gerontology, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yao Zhou
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou, China
| | - Weiwei Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Lu Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Buhui Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Jingjing Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yiye Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Enchao Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Minjie Sun
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Kun Gao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
78
|
Biological Rationale for Targeting MEK/ERK Pathways in Anti-Cancer Therapy and to Potentiate Tumour Responses to Radiation. Int J Mol Sci 2019; 20:ijms20102530. [PMID: 31126017 PMCID: PMC6567863 DOI: 10.3390/ijms20102530] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/16/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
ERK1 and ERK2 (ERKs), two extracellular regulated kinases (ERK1/2), are evolutionary-conserved and ubiquitous serine-threonine kinases involved in regulating cell signalling in normal and pathological tissues. The expression levels of these kinases are almost always different, with ERK2 being the more prominent. ERK1/2 activation is fundamental for the development and progression of cancer. Since their discovery, much research has been dedicated to their role in mitogen-activated protein kinases (MAPK) pathway signalling and in their activation by mitogens and mutated RAF or RAS in cancer cells. In order to gain a better understanding of the role of ERK1/2 in MAPK pathway signalling, many studies have been aimed at characterizing ERK1/2 splicing isoforms, mutants, substrates and partners. In this review, we highlight the differences between ERK1 and ERK2 without completely discarding the hypothesis that ERK1 and ERK2 exhibit functional redundancy. The main goal of this review is to shed light on the role of ERK1/2 in targeted therapy and radiotherapy and highlight the importance of identifying ERK inhibitors that may overcome acquired resistance. This is a highly relevant therapeutic issue that needs to be addressed to combat tumours that rely on constitutively active RAF and RAS mutants and the MAPK pathway.
Collapse
|
79
|
miR-141-5p regulate ATF2 via effecting MAPK1/ERK2 signaling to promote preeclampsia. Biomed Pharmacother 2019; 115:108953. [PMID: 31075732 DOI: 10.1016/j.biopha.2019.108953] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE Preeclampsia is a pregnancy-specific syndrome characterized by hypertension and proteinuria. Impaired trophoblast invasion partly modulated by abnormal MAPK1/ERK2 signaling played important roles in the pathological process of preeclampsia. The objective of this study is to investigate miR-141-5p regulate ATF2 via effecting MAPK1/ERK2 signaling to promote preeclampsia. STUDY DESIGN The maternal placentae and clinical data of 30 patients with preeclampsia and 30 healthy pregnant women were collected in the Second Hospital of Shanxi Medical University from July 2015 to April 2016. Transcriptional levels of miR-141-5p in placentae were monitored using quantitative real-time reverse transcription-polymerase chain reaction. The target gene of miR-141-5p was analyzed with "TargetScanHuman Release 7.2″. To evaluate the pathways of this response, MAPK1 and ERK1/2 in placentae were detected using immunohistochemistry and Western Blot. Transfection experiment was used to verify the function of miR-141-5p regulating ATF2 to effect MAPK1/ERK2 signaling in JEG-3 cells. RESULTS miR-141-5p was significantly down-regulated in placentae of patients with preeclampsia, in comparison to the healthy pregnant women groups. There was no difference in MAPK1 expression between placentae of patients with preeclampsia and healthy pregnant women groups. While p-MAPK1 expression was lower in preeclampsia placentae, in comparison to the healthy pregnant women groups. Moreover, inhibition and activation experiments also validate the function of miR-141-5p in effecting p-MAPK1 level in JEG-3 cells. Bioinformatic analysis identified that ATF2 was a target gene of miR-141-5p, which was one DNA-binding protein to effect phosphatase DUSP1 transcription. DUSP1 effect MAPK1/ERK2 signaling in preeclampsia. CONCLUSION miR-141-5p up-regulated transcription factor ATF2 to promote phosphatase DUSP1 expression. DUSP1 expression reduces p-MAPK1 and ERK1/2 expression to promote preeclampsia.
Collapse
|
80
|
Hausott B, Klimaschewski L. Promotion of Peripheral Nerve Regeneration by Stimulation of the Extracellular Signal-Regulated Kinase (ERK) Pathway. Anat Rec (Hoboken) 2019; 302:1261-1267. [PMID: 30951263 PMCID: PMC6767477 DOI: 10.1002/ar.24126] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/06/2018] [Accepted: 01/11/2019] [Indexed: 12/19/2022]
Abstract
Peripherally projecting neurons undergo significant morphological changes during development and regeneration. This neuroplasticity is controlled by growth factors, which bind specific membrane bound kinase receptors that in turn activate two major intracellular signal transduction cascades. Besides the PI3 kinase/AKT pathway, activated extracellular signal‐regulated kinase (ERK) plays a key role in regulating the mode and speed of peripheral axon outgrowth in the adult stage. Cell culture studies and animal models revealed that ERK signaling is mainly involved in elongative axon growth in vitro and long‐distance nerve regeneration in vivo. Here, we review ERK dependent morphological plasticity in adult peripheral neurons and evaluate the therapeutic potential of interfering with regulators of ERK signaling to promote nerve regeneration. Anat Rec, 302:1261–1267, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Barbara Hausott
- Department of Anatomy, Histology and Embryology, Division of Neuroanatomy, Medical University Innsbruck, Innsbruck, Austria
| | - Lars Klimaschewski
- Department of Anatomy, Histology and Embryology, Division of Neuroanatomy, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
81
|
Williams JA. Cholecystokinin (CCK) Regulation of Pancreatic Acinar Cells: Physiological Actions and Signal Transduction Mechanisms. Compr Physiol 2019; 9:535-564. [PMID: 30873601 DOI: 10.1002/cphy.c180014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreatic acinar cells synthesize and secrete about 20 digestive enzymes and ancillary proteins with the processes that match the supply of these enzymes to their need in digestion being regulated by a number of hormones (CCK, secretin and insulin), neurotransmitters (acetylcholine and VIP) and growth factors (EGF and IGF). Of these regulators, one of the most important and best studied is the gastrointestinal hormone, cholecystokinin (CCK). Furthermore, the acinar cell has become a model for seven transmembrane, heterotrimeric G protein coupled receptors to regulate multiple processes by distinct signal transduction cascades. In this review, we briefly describe the chemistry and physiology of CCK and then consider the major physiological effects of CCK on pancreatic acinar cells. The majority of the review is devoted to the physiologic signaling pathways activated by CCK receptors and heterotrimeric G proteins and the functions they affect. The pathways covered include the traditional second messenger pathways PLC-IP3-Ca2+ , DAG-PKC, and AC-cAMP-PKA/EPAC that primarily relate to secretion. Then there are the protein-protein interaction pathways Akt-mTOR-S6K, the three major MAPK pathways (ERK, JNK, and p38 MAPK), and Ca2+ -calcineurin-NFAT pathways that primarily regulate non-secretory processes including biosynthesis and growth, and several miscellaneous pathways that include the Rho family small G proteins, PKD, FAK, and Src that may regulate both secretory and nonsecretory processes but are not as well understood. © 2019 American Physiological Society. Compr Physiol 9:535-564, 2019.
Collapse
Affiliation(s)
- John A Williams
- University of Michigan, Departments of Molecular & Integrative Physiology and Internal Medicine (Gastroenterology), Ann Arbor, Michigan, USA
| |
Collapse
|
82
|
Ma X, Peng S, Zhou X, Li S, Jin P. The amphioxus ERK2 gene is involved in innate immune response to LPS stimulation. FISH & SHELLFISH IMMUNOLOGY 2019; 86:64-69. [PMID: 30439498 DOI: 10.1016/j.fsi.2018.11.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 06/09/2023]
Abstract
The ERK2 gene is a member of the MAPK family, which plays very important roles in responses to external environmental pressures. However, the ERK2 has yet not been identified in amphioxus to date. To further illuminate the function and evolutionary mechanism of the ERK2 gene, in this present study, we have cloned the full length of the ERK2 gene of Branchiostoma belcheri (designed as AmphiERK2), which is highly homologous to these vertebrate ERK2 genes. The AmphiERK2 protein contains the conserved S_TKc domain and the TEY motif, and its 3D structure is also highly similar to human ERK2 protein. Taken together, our results indicate that the AmphiERK2 gene belongs to a member of the ERK2 gene family. We further use qRT-PCR technology to detect an ubiquitous expression of AmphiERK2 gene in all five investigated tissues (muscle, notochord, gill, hepatic caecum and intestine), and the expression level of AmphiERK2 in both notochord and muscle is significantly higher than the other three tissues. Meanwhile our results also demonstrate that LPS stimulation can induce the up-regulation expression of AmphiERK2 gene and significantly increase the phosphorylation level of AmphiERK2 protein, which seems to imply that the AmphiERK2 may be involved in amphioxus innate immune responses. Overall, our findings provide an important insight into amphioxus innate immune function and evolution of the ERK2 gene family.
Collapse
Affiliation(s)
- Xiangyu Ma
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, 211171, China; The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Shuangli Peng
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Xue Zhou
- School of Chemistry and Biological Engineering, Nanjing Normal University Taizhou College, Taizhou, 225300, China
| | - Shengjie Li
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, 211171, China.
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China.
| |
Collapse
|
83
|
Wang YS, Wu L. Enhanced expression of son of sevenless homolog 1 is predictive of poor prognosis in uveal malignant melanoma patients. Ophthalmic Genet 2019; 40:22-28. [PMID: 30714452 DOI: 10.1080/13816810.2019.1573904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Yan-Shuang Wang
- Department of Ophthalmology, Daqing Oilfield General Hospital, Daqing, Heilongjiang Province, China
| | - Lan Wu
- Department of Ophthalmology, Daqing Oilfield General Hospital, Daqing, Heilongjiang Province, China
| |
Collapse
|
84
|
Liu B, Guo Z, Gao W. miR-181b-5p promotes proliferation and inhibits apoptosis of hypertrophic scar fibroblasts through regulating the MEK/ERK/p21 pathway. Exp Ther Med 2019; 17:1537-1544. [PMID: 30783419 PMCID: PMC6364240 DOI: 10.3892/etm.2019.7159] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/26/2018] [Indexed: 12/22/2022] Open
Abstract
Hypertrophic scar (HS) is a common skin disorder occurring during the wound healing process, and the pathogenesis of HS remains unclear. Previous studies indicated that miRNAs may be involved in the onset and progression of HS. In the present study, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were used to investigate the expression of miR-181b-5p and decorin in HS tissues. Direct interaction between miR-181b-5p and decorin was confirmed using a dual-luciferase assay. Human HS fibroblasts (HSFbs) were cultured and transfected with miR-181b-5p mimics, and MTT assay and Annexin V fluorescein isothiocyanate/propidium iodide staining were performed to investigate the role of miR-181b-5p in the proliferation and apoptosis of HSFbs. Subsequently, the expression levels of mitogen-activated protein kinase kinase (MEK), phospho (p)-extracellular signal-regulated kinase (ERK) and p21 were determined in HSFbs transfected with miR-181b-5p mimics and untransfected cells using RT-qPCR and western blotting. The results indicated upregulation of miR-181b-5p and downregulation of decorin expression in HS tissues compared with normal skin samples. miR-181b-5p may regulate the expression of decorin through direct binding to the 3′-untranslated region, as demonstrated by the results of the dual-luciferase assay. Transfection with miR-181b-5p mimics in HSFbs enhanced cell proliferation, reduced apoptosis and increased the expression of MEK, p-ERK and p21. Furthermore, treatment with MEK inhibitor in HSFbs transfected with miR-181b-5p mimics partially inhibited miR-181b-5p-induced antiapoptotic effects. Taken together, increased expression of miR-181b-5p may serve important roles in the pathogenesis of HS through regulating the MEK/ERK/p21 pathway, suggesting that miR-181b-5p may be a therapeutic target for the treatment of HS.
Collapse
Affiliation(s)
- Bo Liu
- Department of Medical Cosmetology, Eastern Liaoning University, Dandong, Liaoning 118003, P.R. China
| | - Zhe Guo
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Weiming Gao
- Department of Medicine, Eastern Liaoning University, Dandong, Liaoning 118003, P.R. China
| |
Collapse
|
85
|
Zhang C, Zhou J, Hu J, Lei S, Yuan M, Chen L, Wang G, Qiu Z. Celecoxib attenuates hepatocellular proliferative capacity during hepatocarcinogenesis by modulating a PTEN/NF-κB/PRL-3 pathway. RSC Adv 2019; 9:20624-20632. [PMID: 35515542 PMCID: PMC9065693 DOI: 10.1039/c9ra00429g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 06/01/2019] [Indexed: 11/21/2022] Open
Abstract
Although the efficacy of celecoxib on various cancer cell behaviors, including aberrant proliferation, in cultured hepatocellular carcinoma (HCC) cells has been demonstrated, whether celecoxib regulates cell proliferation by targeting PRL-3-associated signaling transduction during hepatocarcinogenesis in vivo has been incompletely studied. Here, we investigate the anti-proliferative efficacy of celecoxib in a rapid HCC mouse model established by hydrodynamic transfection of activated AKT and c-Met proto-oncogenes. The results show that celecoxib is effective at delaying the malignant transformation of hepatocytes by reducing the protein expression of Ki67, Cyclin D1 and c-Myc in the AKT/c-Met HCC-bearing mice. Mechanistically, celecoxib increases the protein expression of PTEN and suppresses the protein expression of NF-κB and PRL-3 in the liver of the HCC mice. Using PTEN-silenced and LPS-stimulated approaches in vitro, a mechanism by which celecoxib regulates a PTEN/NF-κB/PRL-3 pathway in HCC cells was illuminated. Altogether, our study demonstrates that celecoxib attenuates the hepatocellular proliferative capacity during hepatocarcinogenesis, which is probably attributable to its regulation of the PTEN/NF-κB/PRL-3 pathway. Celecoxib modulates the PTEN/NF-κB/PRL-3 pathway during hepatocarcinogenesis in vivo.![]()
Collapse
Affiliation(s)
- Cong Zhang
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Junxuan Zhou
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Junjie Hu
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Sheng Lei
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Ming Yuan
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Liang Chen
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Guihong Wang
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription
| | - Zhenpeng Qiu
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
- Key Laboratory of Resources and Chemistry of Chinese Medicine
| |
Collapse
|
86
|
Vassilakos G, Barton ER. Insulin-Like Growth Factor I Regulation and Its Actions in Skeletal Muscle. Compr Physiol 2018; 9:413-438. [PMID: 30549022 DOI: 10.1002/cphy.c180010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The insulin-like growth factor (IGF) pathway is essential for promoting growth and survival of virtually all tissues. It bears high homology to its related protein insulin, and as such, there is an interplay between these molecules with regard to their anabolic and metabolic functions. Skeletal muscle produces a significant proportion of IGF-1, and is highly responsive to its actions, including increased muscle mass and improved regenerative capacity. In this overview, the regulation of IGF-1 production, stability, and activity in skeletal muscle will be described. Second, the physiological significance of the forms of IGF-1 produced will be discussed. Last, the interaction of IGF-1 with other pathways will be addressed. © 2019 American Physiological Society. Compr Physiol 9:413-438, 2019.
Collapse
Affiliation(s)
- Georgios Vassilakos
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
87
|
Zhang F, Cao H. MicroRNA‑143‑3p suppresses cell growth and invasion in laryngeal squamous cell carcinoma via targeting the k‑Ras/Raf/MEK/ERK signaling pathway. Int J Oncol 2018; 54:689-701. [PMID: 30535502 DOI: 10.3892/ijo.2018.4655] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 07/02/2018] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) have been identified as an important regulator in carcinogenesis and other pathological processes. However, the molecular mechanism underlying the function of miRNAs in the progression and development of laryngeal squamous cell carcinoma (LSCC) remains to be fully elucidated. In the present study, the miRNA expression pattern in LSCC tissues was profiled using miRNA microarray analysis. It was found that a large set of miRNAs are aberrantly expressed in LSCC tissues and that miR‑143‑3p was the most markedly downregulated compared with normal tissues. The low expression of miR‑143‑3p was associated with poor prognosis in LSCC. The overexpression of miR‑143‑3p repressed cellular proliferation and induced apoptosis in vitro, and inhibited tumor growth in vivo. The upregulation of miR‑143‑3p suppressed cell migration and invasion through inhibiting the epithelial‑mesenchymal transition cascade. In addition, it was verified that the oncogene k‑Ras is a target of miR‑143‑3p in LSCC cells, and the suppressive effects of miR‑143‑3p on LSCC cells were abrogated by the overexpression of k‑Ras. It was also revealed that miR‑143‑3p may inhibit cell growth and metastasis through targeting the k‑Ras/Raf/mitogen‑activated protein kinase kinase (MEK)/extracellular signal‑regulated kinase (ERK) signaling pathway. Taken together, the data indicated that the miR‑143‑3p/k‑Ras/Raf/MEK/ERK axis serves a key regulator in the development and progression of LSCC, suggesting that miR‑143‑3p may be a potential prognostic biomarker and therapeutic target in the treatment of LSCC.
Collapse
Affiliation(s)
- Feng Zhang
- Ear Nose and Throat Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hua Cao
- Ear Nose and Throat Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
88
|
Nahari E, Razi M. Silymarin amplifies apoptosis in ectopic endometrial tissue in rats with endometriosis; implication on growth factor GDNF, ERK1/2 and Bcl-6b expression. Acta Histochem 2018; 120:757-767. [PMID: 30195499 DOI: 10.1016/j.acthis.2018.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/29/2018] [Accepted: 08/16/2018] [Indexed: 12/25/2022]
Abstract
The present prospective study was done to evaluate the effect of silymarin (SMN) on endometriotic-like legions establishment and growth in experimentally-induced endometriosis. For this purpose, the experimental endometriosis was induced in 12 rats and then the animals subdivided into endometriosis-sole and SMN (50 mg kg-1, orally)+endometriosis groups. Following 28 days, the legions establishment, size, Glial cell line-derived neurotrophic factor (GDNF), gfrα1, B Cell Lymphoma 6 (Bcl-6b), Bcl-2, extracellular regulator kinase (ERK1/2) expression ratios, angiogenesis, the apoptosis and fibrosis indices were investigated. The SMN significantly (P < 0.05) decreased the enometriotic-like legions establishment and size, decreased mRNA levels of GDNF, gfrα1, Bcl-6b and Bcl-2 and remarkably diminished GDNF, gfrα1, Bcl-6b and Bcl-2-positive cells distribution/mm2 of tissue versus endometriosis-sole group. The SMN + endometriosis group exhibited a significant (P < 0.05) enhancement in ERK1/2 expression and represented diminished vascularized area and increased apoptosis and fibrosis indices, as well. In conclusion, the SMN by down-regulating GDNF and its receptor gfrα1 expression inhibits GDNF-gfrα1 complex generation and consequently suppresses Bcl-6b expression. Moreover, the SMN by enhancing the ERK1/2 expression and by suppressing the Bcl-2 expression promotes the apoptosis pathway. Finally, the SMN by down-regulating the angiogenesis ratio accelerates apoptosis and consequently induces severe fibrosis in endometriotic-like legions.
Collapse
Affiliation(s)
- Elaheh Nahari
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Mazdak Razi
- Department of Basic Sciences, Faculty of Veterinary Medicine, P.O. BOC: 1177, Urmia University, Urmia, Iran.
| |
Collapse
|
89
|
α-Hederin Arrests Cell Cycle at G2/M Checkpoint and Promotes Mitochondrial Apoptosis by Blocking Nuclear Factor-κB Signaling in Colon Cancer Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2548378. [PMID: 30363706 PMCID: PMC6180961 DOI: 10.1155/2018/2548378] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/08/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022]
Abstract
Colon cancer represents the third most common malignancy worldwide. New drugs with high efficaciousness and safety for the treatment of colon cancer are urgently needed in clinical context. Here, we were aimed to evaluate the antitumor activity of the natural compound α-hederin in human colon cancer cells. We treated SW620 cells with interleukin-6 (IL-6) in vitro to mimic the paracrine inflammatory microenvironment of tumor cells. α-Hederin concentration dependently reduced the viability of IL-6-stimulated SW620 cells. α-Hederin increased the number of IL-6-stimulated SW620 cells at the G2/M phase and reduced the mRNA and protein expression of cyclin B1 and CDK1. Moreover, α-hederin induced apoptosis and loss of mitochondrial membrane potential in IL-6-stimulated SW620 cells. α-Hederin downregulated Bcl-2 expression, upregulated Bax expression, and promoted cytochrome c release from mitochondria into cytoplasm. Additionally, α-hederin elevated the levels of cleaved-caspase-9, cleaved-caspase-3, and cleaved-PARP, but had little effects on the levels of cleaved-caspase-8. Moreover, α-hederin prevented the nuclear translocation of nuclear factor-κB (NF-κB) and reduced the phosphorylation of IκBα and IKKα, suggesting the blockade of NF-κB signaling. NF-κB inhibitor PDTC not only produced similar proapoptotic effects on IL-6-stimulated SW620 cells as α-hederin did, but also synergistically enhanced α-hederin's proapoptotic effects. Furthermore, α-hederin inhibited the phosphorylation of ERK in IL-6-stimulated SW620 cells, which was involved in α-hederin blockade of NF-κB nuclear translocation. Altogether, α-hederin suppressed viability, induced G2/M cell cycle arrest, and stimulated mitochondrial and caspase-dependent apoptosis in colon cancer cells, which were associated with disruption of NF-κB and ERK pathways, suggesting α-hederin as a promising candidate for intervention of colon cancer.
Collapse
|
90
|
Focal Adhesion Kinase and ROCK Signaling Are Switch-Like Regulators of Human Adipose Stem Cell Differentiation towards Osteogenic and Adipogenic Lineages. Stem Cells Int 2018; 2018:2190657. [PMID: 30275837 PMCID: PMC6157106 DOI: 10.1155/2018/2190657] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/07/2018] [Accepted: 07/04/2018] [Indexed: 12/18/2022] Open
Abstract
Adipose tissue is an attractive stem cell source for soft and bone tissue engineering applications and stem cell therapies. The adipose-derived stromal/stem cells (ASCs) have a multilineage differentiation capacity that is regulated through extracellular signals. The cellular events related to cell adhesion and cytoskeleton have been suggested as central regulators of differentiation fate decision. However, the detailed knowledge of these molecular mechanisms in human ASCs remains limited. This study examined the significance of focal adhesion kinase (FAK), Rho-Rho-associated protein kinase (Rho-ROCK), and their downstream target extracellular signal-regulated kinase 1/2 (ERK1/2) on hASCs differentiation towards osteoblasts and adipocytes. Analyses of osteogenic markers RUNX2A, alkaline phosphatase, and matrix mineralization revealed an essential role of active FAK, ROCK, and ERK1/2 signaling for the osteogenesis of hASCs. Inhibition of these kinases with specific small molecule inhibitors diminished osteogenesis, while inhibition of FAK and ROCK activity led to elevation of adipogenic marker genes AP2 and LEP and lipid accumulation implicating adipogenesis. This denotes to a switch-like function of FAK and ROCK signaling in the osteogenic and adipogenic fates of hASCs. On the contrary, inhibition of ERK1/2 kinase activity deceased adipogenic differentiation, indicating that activation of ERK signaling is required for both adipogenic and osteogenic potential. Our findings highlight the reciprocal role of cell adhesion mechanisms and actin dynamics in regulation of hASC lineage commitment. This study enhances the knowledge of molecular mechanisms dictating hASC differentiation and thus opens possibilities for more efficient control of hASC differentiation.
Collapse
|
91
|
Oyagbemi AA, Omobowale TO, Ola-Davies OE, Asenuga ER, Ajibade TO, Adejumobi OA, Arojojoye OA, Afolabi JM, Ogunpolu BS, Falayi OO, Hassan FO, Ochigbo GO, Saba AB, Adedapo AA, Yakubu MA. Quercetin attenuates hypertension induced by sodium fluoride via reduction in oxidative stress and modulation of HSP 70/ERK/PPARγ signaling pathways. Biofactors 2018; 44:465-479. [PMID: 30171731 DOI: 10.1002/biof.1445] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
Hypertension is one of the silent killers in the world with high mortality and morbidity. The exposure of humans and animals to fluoride and/or fluoride containing compounds is almost inevitable. This study investigated the modulatory effects of quercetin on sodium fluoride (NaF)-induced hypertension and cardiovascular complications. Forty male rats were randomly separated into four groups (n =10). Group A animals served as the control, rats in Group B were exposed to 300 ppm of NaF, Groups C and D animals were exposed to 300 ppm of NaF along with quercetin orally at 50 mg/kg and 100 mg/kg orally by gavage, while NaF was administered in drinking water, respectively, for a week. Administration of NaF caused severe hypertension as indicated with significant increases in the systolic, diastolic, and mean arterial blood pressure, together with prolonged ventricular depolarization (QRS) and the time between the start of the Q wave and the end of the T wave in the heart's electrical cycle (QT) intervals when compared with controls. NaF significantly decreased the activities of antioxidant enzymes, caused increase in markers of oxidative stress and renal damage when compared with controls. Immunohistochemical staining revealed lower expressions of Hsp70, ERK, and PPARγ in the heart, kidney, and aorta of rats-administered NaF relative to the controls. Together, quercetin co-treatment with NaF restored blood pressure, normalized QRS interval, and improved antioxidant defense system. © 2018 BioFactors, 44(5):465-479, 2018.
Collapse
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temidayo Olutayo Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Eunice Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ebunoluwa Racheal Asenuga
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Benin, Benin, Nigeria
| | - Temitayo Olabisi Ajibade
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumuyiwa Abiola Adejumobi
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Jeremiah Moyinoluwa Afolabi
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
- Cell Biology & Physiology track, Integrated Biomedical Sciences PhD, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Blessing Seun Ogunpolu
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Olubunmi Falayi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Fasilat Oluwakemi Hassan
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Grace Onyeche Ochigbo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adebowale Benard Saba
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeolu Alex Adedapo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Momoh Audu Yakubu
- Department of Environmental and Interdisciplinary Sciences, College of Science, Engineering and Technology, Vascular Biology Unit, Center for Cardiovascular Diseases, COPHS, Texas Southern University, Houston, TX, USA
| |
Collapse
|
92
|
Zhang M, Qian C, Zheng ZG, Qian F, Wang Y, Thu PM, Zhang X, Zhou Y, Tu L, Liu Q, Li HJ, Yang H, Li P, Xu X. Jujuboside A promotes Aβ clearance and ameliorates cognitive deficiency in Alzheimer's disease through activating Axl/HSP90/PPARγ pathway. Theranostics 2018; 8:4262-4278. [PMID: 30128052 PMCID: PMC6096387 DOI: 10.7150/thno.26164] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/03/2018] [Indexed: 01/20/2023] Open
Abstract
Rationale: It has been reported that peroxisome proliferator activated receptor γ (PPARγ) level decreases significantly in the brains of Alzheimer's disease (AD) patients and mice models, while the mechanism is unclear. This study aims to unravel the mechanism that amyloid β (Aβ) decreases PPARγ and attempted to discover lead compound that preserves PPARγ. Methods: In APP/PS1 transgenic mice and Aβ treated microglia, the interaction between HSP90 and PPARγ were analyzed by western blot. Using a PPRE (PPARγ responsive element) containing reporter cell line, compounds that activate PPARγ activity were identified. After genetic ablation or pharmacological inhibition of potential target pathways, the target of jujuboside A (JuA) was discovered through Axl/HSP90β. After oral administration or intrathecal injection, the anti-AD activity of JuA was evaluated by Morris water maze (MWM) test and object recognition test. Soluble Aβ42 levels and plaque numbers after JuA treatment were detected by thioflavin S staining, and the activation of microglia was assayed by immunofluorescence staining against Iba-1. Results: We found that Aβ stress decreased heat shock protein 90 β (HSP90β), subsequently reduced the abundance of PPARγ, and down-regulated Aβ clearance-related genes in BV2 cells and primary microglia. We identified that JuA stimulated the expression of HSP90β, strengthened the interaction between HSP90β and PPARγ, preserved PPARγ levels, and thus effectively promoted the clearance of Aβ42. We demonstrated that JuA increased HSP90β expression through Axl/ERK pathway. JuA significantly ameliorated cognitive deficiency in APP/PS1 transgenic mice, meanwhile, JuA significantly reduced the soluble Aβ42 levels and plaque numbers in the brain. Notably, the therapeutic effects of JuA were dampened by R428, an Axl inhibitor. Conclusions: This study suggests that the up-regulation of HSP90β by JuA through Axl is a potential therapeutic strategy to facilitate Aβ42 clearance and ameliorate cognitive deficiency in AD.
Collapse
Affiliation(s)
- Mu Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Cheng Qian
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Fei Qian
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Yanyan Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Pyone Myat Thu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Yaping Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Lifan Tu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Qingling Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Hua Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Metabolic Disease, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| |
Collapse
|
93
|
Elucidating molecular events underlying topography mediated cardiomyogenesis of stem cells on 3D nanofibrous scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 88:104-114. [DOI: 10.1016/j.msec.2018.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/27/2018] [Accepted: 03/14/2018] [Indexed: 12/28/2022]
|
94
|
Liu F, Yang X, Geng M, Huang M. Targeting ERK, an Achilles' Heel of the MAPK pathway, in cancer therapy. Acta Pharm Sin B 2018; 8:552-562. [PMID: 30109180 PMCID: PMC6089851 DOI: 10.1016/j.apsb.2018.01.008] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/11/2017] [Accepted: 01/08/2018] [Indexed: 12/11/2022] Open
Abstract
The mitogen-activated protein kinases (MAPK) pathway, often known as the RAS-RAF-MEK-ERK signal cascade, functions to transmit upstream signals to its downstream effectors to regulate physiological process such as cell proliferation, differentiation, survival and death. As the most frequently mutated signaling pathway in human cancer, targeting the MAPK pathway has long been considered a promising strategy for cancer therapy. Substantial efforts in the past decades have led to the clinical success of BRAF and MEK inhibitors. However, the clinical benefits of these inhibitors are compromised by the frequently occurring acquired resistance due to cancer heterogeneity and genomic instability. This review briefly introduces the key protein kinases involved in this pathway as well as their activation mechanisms. We also generalize the correlations between mutations of MAPK members and human cancers, followed by a summarization of progress made on the development of small molecule MAPK kinases inhibitors. In particular, this review highlights the potential advantages of ERK inhibitors in overcoming resistance to upstream targets and proposes that targeting ERK kinase may hold a promising prospect for cancer therapy.
Collapse
|
95
|
Qi L, Jiang J, Jin P, Kuang M, Wei Q, Shi F, Mao D. Expression patterns of claudin-5 and its related signals during luteal regression in pseudopregnant rats: The enhanced effect of additional PGF treatment. Acta Histochem 2018; 120:221-227. [PMID: 29449022 DOI: 10.1016/j.acthis.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/29/2018] [Accepted: 02/06/2018] [Indexed: 12/23/2022]
Abstract
To study the expression patterns of claudin-5 and its related signals during luteal regression in rats, a sequential PMSG/hCG treatment paradigm was used to obtain a single, well-defined generation of corpus luteum (CL). A total of 35 rats were treated with one PGF or two PGF at an interval of 24 h from day 7 of pseudopregnancy to induce CL regression. Serum and ovaries were collected at 0, 2, 4, 8 or 24 h after one PGF injection (1 PGF), 2 or 24 h after two PGF injections (2 PGF). The serum progesterone level was detected by RIA; the ovarian expression of claudin-5, the phosphorylations of STAT3 (p-STAT3), Akt (p-Akt), ERK1/2 (p-ERK) and p38 MAPK (p-p38) were detected by western blot, real-time PCR and IHC. Results showed that serum progesterone (P4) decreased after PGF treatment. Claudin-5 mRNA decreased at 4 h and 8 h after 1 PGF and 2 h after 2 PGF, and claudin-5 protein decreased at 4 h after 1 PGF. p-STAT3 increased at 4 h after 1 PGF and 2 h after 2 PGF. p-ERK increased at 2 h after 2 PGF. The level of p-Akt decreased at 4 h after 1 PGF. PGF treatment did not alter the phosphorylation of p38 MAPK at any time points in this study. IHC results revealed that claudin-5 was expressed in the nuclei and cytoplasm of steroidogenic cells and in the vessels, while PGF induced-p-STAT3 was expressed uniformly in the cytoplasm of luteal steroidogenic cells. In conclusion, PGF treatment decreased the expression of claudin-5 and the additional PGF treatment enhanced the decrease in claudin-5 mRNA expression and the increases in ERK1/2 and STAT3 phosphorylation in the corpus luteum of pseudopregnant rats, which will contribute new information to the further study of molecular mechanism of luteal regression.
Collapse
|
96
|
Stelma T, Leaner VD. KPNB1-mediated nuclear import is required for motility and inflammatory transcription factor activity in cervical cancer cells. Oncotarget 2018; 8:32833-32847. [PMID: 28427184 PMCID: PMC5464831 DOI: 10.18632/oncotarget.15834] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/07/2017] [Indexed: 12/14/2022] Open
Abstract
Karyopherin β1 is a nuclear import protein involved in the transport of proteins containing a nuclear localisation sequence. Elevated Karyopherin β1 expression has been reported in cancer and transformed cells and is essential for cancer cell proliferation and survival. Transcription factors such as NFĸB and AP-1 contain a nuclear localisation sequence and initiate the expression of multiple factors associated with inflammation and cancer cell biology. Our study investigated the effect of inhibiting nuclear import via Karyopherin β1 on cancer cell motility and inflammatory signaling using siRNA and the novel small molecule, Inhibitor of Nuclear Import-43, INI-43. Inhibition of Karyopherin β1 led to reduced migration and invasion of cervical cancer cells. Karyopherin β1 is essential for the translocation of NFĸB into the nucleus as nuclear import inhibition caused its cytoplasmic retention and decreased transcriptional activity. A similar decrease was seen in AP-1 transcriptional activity upon Karyopherin β1 inhibition. Consequently reduced interleukin-6, interleukin-1 beta, tumour necrosis factor alpha and granulocyte macrophage colony stimulating factor expression, target genes of NFkB and AP-1, was observed. Migration studies inhibiting individual transcription factors suggested that INI-43 may affect a combination of signaling events. Our study provides further evidence that inhibiting KPNB1 has anti-cancer effects and shows promise as a chemotherapeutic target.
Collapse
Affiliation(s)
- Tamara Stelma
- Division of Medical Biochemistry and Structural Biology, SAMRC Gynaecology Cancer Research Centre, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Virna D Leaner
- Division of Medical Biochemistry and Structural Biology, SAMRC Gynaecology Cancer Research Centre, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
97
|
RSK2 activity mediates glioblastoma invasiveness and is a potential target for new therapeutics. Oncotarget 2018; 7:79869-79884. [PMID: 27829215 PMCID: PMC5346757 DOI: 10.18632/oncotarget.13084] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 10/21/2016] [Indexed: 01/06/2023] Open
Abstract
In glioblastoma (GBM), infiltration of primary tumor cells into the normal tissue and dispersal throughout the brain is a central challenge to successful treatment that remains unmet. Indeed, patients respond poorly to the current therapies of tumor resection followed by chemotherapy with radiotherapy and have only a 16-month median survival. It is therefore imperative to develop novel therapies. RSK2 is a kinase that regulates proliferation and adhesion and can promote metastasis. We demonstrate that active RSK2 regulates GBM cell adhesion and is essential for cell motility and invasion of patient-derived GBM neurospheres. RSK2 control of adhesion and migration is mediated in part by its effects on integrin-Filamin A complexes. Importantly, inhibition of RSK2 by either RSK inhibitors or shRNA silencing impairs invasion and combining RSK2 inhibitors with temozolomide improves efficacy in vitro. In agreement with the in vitro data, using public datasets, we find that RSK2 is significantly upregulated in vivo in human GBM patient tumors, and that high RSK2 expression significantly correlates with advanced tumor stage and poor patient survival. Together, our data provide strong evidence that RSK inhibitors could enhance the effectiveness of existing GBM treatment, and support RSK2 targeting as a promising approach for novel GBM therapy.
Collapse
|
98
|
Gentile MT, Russo R, Pastorino O, Cioffi S, Barbieri F, Illingworth EA, Grieco M, Chambery A, Colucci-D'Amato L. Ruta graveolens water extract inhibits cell-cell network formation in human umbilical endothelial cells via MEK-ERK1/2 pathway. Exp Cell Res 2018; 364:50-58. [PMID: 29366810 DOI: 10.1016/j.yexcr.2018.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 01/10/2018] [Accepted: 01/19/2018] [Indexed: 01/01/2023]
Abstract
Angiogenesis is a process encompassing several steps such as endothelial cells proliferation, differentiation and migration to form a vascular network, involving different signal transduction pathways. Among these, ERK1/2 signaling mediates VEGF-dependent signaling pathway. Here we report that the water extract of Ruta graveolens (RGWE), widely known as a medicinal plant, is able to impair in a dose-dependent manner, cell network formation without affecting cell viability. Biochemical analysis showed that the major component of RGWE is rutin, unable to reproduce RGWE effect. We found that RGWE inhibits ERK1/2 phosphorylation and that this event is crucial in cell network formation since the transfection of HUVEC with a constitutively active MEK (caMEK), the ERK1/2 activator, induces a robust cell network formation as compared to untransfected and/or mock transfected cells and, more importantly, caMEK transfected cells became unresponsive to RGWE. Moreover, RGWE inhibits VEGF and nestin gene expression, necessary for vessel formation, and the caMEK transfection induces their higher expression. In conclusion, we report that RGWE is able to significantly impair vessels network formation without affecting cell viability, preventing ERK1/2 activation and, in turn, down-regulating VEGF and nestin expression. These findings point to RGWE as a potential therapeutic tool capable to interfere with pathologic angiogenesis.
Collapse
Affiliation(s)
- Maria Teresa Gentile
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Rosita Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Olga Pastorino
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Sara Cioffi
- Dipartimento di Chimica e Biologia "Adolfo Zambelli", Università degli Studi di Salerno, Italy; Institute of Genetics and Biophysics "ABT", CNR, Naples, Italy
| | | | - Elisabeth Anne Illingworth
- Dipartimento di Chimica e Biologia "Adolfo Zambelli", Università degli Studi di Salerno, Italy; Institute of Genetics and Biophysics "ABT", CNR, Naples, Italy
| | - Michele Grieco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Luca Colucci-D'Amato
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
99
|
Agudo-Ibañez L, Crespo P, Casar B. Analysis of Ras/ERK Compartmentalization by Subcellular Fractionation. Methods Mol Biol 2018; 1487:151-162. [PMID: 27924565 DOI: 10.1007/978-1-4939-6424-6_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
A vast number of stimuli use the Ras/Raf/MEK/ERK signaling cascade to transmit signals from their cognate receptors, in order to regulate multiple cellular functions, including key processes such as proliferation, cell cycle progression, differentiation, and survival. The duration, intensity and specificity of the responses are, in part, controlled by the compartmentalization/subcellular localization of the signaling intermediaries. Ras proteins are found in different plasma membrane microdomains and endomembranes. At these localizations, Ras is subject to site-specific regulatory mechanisms, distinctively engaging effector pathways and switching-on diverse genetic programs to generate a multitude of biological responses. The Ras effector pathway leading to ERKs activation is also subject to space-related regulatory processes. About half of ERK1/2 substrates are found in the nucleus and function mainly as transcription factors. The other half resides in the cytosol and other cellular organelles. Such subcellular distribution enhances the complexity of the Ras/ERK cascade and constitutes an essential mechanism to endow variability to its signals, which enables their participation in the regulation of a broad variety of functions. Thus, analyzing the subcellular compartmentalization of the members of the Ras/ERK cascade constitutes an important factor to be taken into account when studying specific biological responses evoked by Ras/ERK signals. Herein, we describe methods for such purpose.
Collapse
Affiliation(s)
- Lorena Agudo-Ibañez
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas - Universidad de Cantabria, c/Albert Einstein, 22, PCTCAN, Santander, 39011, Cantabria, Spain
| | - Piero Crespo
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas - Universidad de Cantabria, c/Albert Einstein, 22, PCTCAN, Santander, 39011, Cantabria, Spain
| | - Berta Casar
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas - Universidad de Cantabria, c/Albert Einstein, 22, PCTCAN, Santander, 39011, Cantabria, Spain.
| |
Collapse
|
100
|
Tang F, Pacheco MTF, Chen P, Liang D, Li W. Secretogranin III promotes angiogenesis through MEK/ERK signaling pathway. Biochem Biophys Res Commun 2018; 495:781-786. [PMID: 29154827 PMCID: PMC5736013 DOI: 10.1016/j.bbrc.2017.11.080] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/11/2017] [Indexed: 01/08/2023]
Abstract
Secretogranin III (Scg3) was recently discovered as the first highly diabetic retinopathy-associated angiogenic factor, and its neutralizing antibody alleviated the disease with high efficacy in diabetic mice. Investigation of its molecular mechanisms will facilitate the translation of this novel therapy. Scg3 was reported to induce the phosphorylation of mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK). Here we characterized the importance of MEK/ERK activation to Scg3 angiogenic activity. Our results showed that MEK inhibitor PD98059 blocked Scg3-induced proliferation of human umbilical vein endothelial cells (HUVECs). This finding was corroborated by PD98059 inhibition of HUVEC migration and tube formation. Furthermore, ERK inhibitor SCH772984 also suppressed Scg3-induced proliferation and migration of HUVECs. Taken together, these findings suggest that MEK-ERK pathway plays an important role in Scg3-induced angiogenesis.
Collapse
Affiliation(s)
- Fen Tang
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, FL, USA; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Mario Thiego F Pacheco
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, FL, USA
| | - Ping Chen
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, FL, USA; Department of Ophthalmology, Renji Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Dan Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Wei Li
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, FL, USA; Vascular Biology Institute, University of Miami, School of Medicine, Miami, FL, USA.
| |
Collapse
|