201
|
Thaker YR, Raab M, Strebhardt K, Rudd CE. GTPase-activating protein Rasal1 associates with ZAP-70 of the TCR and negatively regulates T-cell tumor immunity. Nat Commun 2019; 10:4804. [PMID: 31641113 PMCID: PMC6805919 DOI: 10.1038/s41467-019-12544-4] [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: 05/20/2018] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy involving checkpoint blockades of inhibitory co-receptors is effective in combating cancer. Despite this, the full range of mediators that inhibit T-cell activation and influence anti-tumor immunity is unclear. Here, we identify the GTPase-activating protein (GAP) Rasal1 as a novel TCR-ZAP-70 binding protein that negatively regulates T-cell activation and tumor immunity. Rasal1 inhibits via two pathways, the binding and inhibition of the kinase domain of ZAP-70, and GAP inhibition of the p21ras-ERK pathway. It is expressed in activated CD4 + and CD8 + T-cells, and inhibits CD4 + T-cell responses to antigenic peptides presented by dendritic cells as well as CD4 + T-cell responses to peptide antigens in vivo. Furthermore, siRNA reduction of Rasal1 expression in T-cells shrinks B16 melanoma and EL-4 lymphoma tumors, concurrent with an increase in CD8 + tumor-infiltrating T-cells expressing granzyme B and interferon γ-1. Our findings identify ZAP-70-associated Rasal1 as a new negative regulator of T-cell activation and tumor immunity. Activation of T cells in the tumor microenvironment can be inhibited through a variety of mechanisms. Here, the authors show that Rasal1, a GTPase-activating protein, binds and inhibits signaling downstream of the T Cell Receptor complex and that consistently, its reduced expression enhances anti-tumor T-cell responses in two syngeneic cancer mouse models.
Collapse
Affiliation(s)
- Youg Raj Thaker
- Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.,School of Biological Science, Protein Structure and Disease Mechanisms, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Monika Raab
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Klaus Strebhardt
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Christopher E Rudd
- Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK. .,Département de Immunologie-Oncologie, Centre de Recherche Hôpital Maisonneuve-Rosemont, Montreal, QC, H1T 2M4, Canada. .,Département de Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
202
|
Baig MH, Baker A, Ashraf GM, Dong JJ. ASK1 and its role in cardiovascular and other disorders: available treatments and future prospects. Expert Rev Proteomics 2019; 16:857-870. [DOI: 10.1080/14789450.2019.1676735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Mohammad Hassan Baig
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| | - Abu Baker
- Nanobiotechnology and nanomedicine lab, Department of Biosciences, Integral University, Lucknow, India
| | - Ghulam M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jae-June Dong
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| |
Collapse
|
203
|
Wang ZH, Liu Y, Chaitankar V, Pirooznia M, Xu H. Electron transport chain biogenesis activated by a JNK-insulin-Myc relay primes mitochondrial inheritance in Drosophila. eLife 2019; 8:49309. [PMID: 31612862 PMCID: PMC6809605 DOI: 10.7554/elife.49309] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/13/2019] [Indexed: 12/25/2022] Open
Abstract
Oogenesis features an enormous increase in mitochondrial mass and mtDNA copy number, which are required to furnish mature eggs with an adequate supply of mitochondria and to curb the transmission of deleterious mtDNA variants. Quiescent in dividing germ cells, mtDNA replication initiates upon oocyte determination in the Drosophila ovary, which necessitates active mitochondrial respiration. However, the underlying mechanism for this dynamic regulation remains unclear. Here, we show that an feedforward insulin-Myc loop promotes mitochondrial respiration and biogenesis by boosting the expression of electron transport chain subunits and of factors essential for mtDNA replication and expression, and for the import of mitochondrial proteins. We further reveal that transient activation of JNK enhances the expression of the insulin receptor and initiates the insulin-Myc signaling loop. This signaling relay promotes mitochondrial biogenesis in the ovary, and thereby plays a role in limiting the transmission of deleterious mtDNA mutations. Our study demonstrates cellular mechanisms that couple mitochondrial biogenesis and inheritance with oocyte development.
Collapse
Affiliation(s)
- Zong-Heng Wang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Yi Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Vijender Chaitankar
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Mehdi Pirooznia
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Hong Xu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| |
Collapse
|
204
|
PPA1 promotes NSCLC progression via a JNK- and TP53-dependent manner. Oncogenesis 2019; 8:53. [PMID: 31551407 PMCID: PMC6760234 DOI: 10.1038/s41389-019-0162-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 08/11/2019] [Accepted: 08/26/2019] [Indexed: 11/16/2022] Open
Abstract
Inorganic pyrophosphatase (PPA1) promotes tumor progression in several tumor types. However, the underlying mechanism remains elusive. Here, we disclosed that PPA1 expression is markedly upregulated in lung carcinoma tissue versus normal lung tissue. We also found that the non-small cell lung cancer (NSCLC) cell lines show increased PPA1 expression levels versus normal lung cell line control. Moreover, the knockdown of PPA1 promotes cell apoptosis and inhibits cell proliferation. Whereas, the ectopic expression of PPA1 reduces cell apoptosis and enhances cell proliferation. Most interestingly, the expression of mutant PPA1 (D117A) significantly abolishes PPA1-mediated effect on cell apoptosis and proliferation. The underlying mechanism demonstrated that TP53 expression deficiency or JNK inhibitor treatment could abolish PPA1-mediated NSCLC progression. In summary, the aforementioned findings in this study suggest a new pathway the PPA1 mediates NSCLC progression either via TP53 or JNK. Most important, the pyrophosphatase activity is indispensible for PPA1-mediated NSCLC progression. This may provide a promising target for NSCLC therapy.
Collapse
|
205
|
Moens U, Macdonald A. Effect of the Large and Small T-Antigens of Human Polyomaviruses on Signaling Pathways. Int J Mol Sci 2019; 20:ijms20163914. [PMID: 31408949 PMCID: PMC6720190 DOI: 10.3390/ijms20163914] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/09/2019] [Accepted: 08/10/2019] [Indexed: 12/12/2022] Open
Abstract
Viruses are intracellular parasites that require a permissive host cell to express the viral genome and to produce new progeny virus particles. However, not all viral infections are productive and some viruses can induce carcinogenesis. Irrespective of the type of infection (productive or neoplastic), viruses hijack the host cell machinery to permit optimal viral replication or to transform the infected cell into a tumor cell. One mechanism viruses employ to reprogram the host cell is through interference with signaling pathways. Polyomaviruses are naked, double-stranded DNA viruses whose genome encodes the regulatory proteins large T-antigen and small t-antigen, and structural proteins that form the capsid. The large T-antigens and small t-antigens can interfere with several host signaling pathways. In this case, we review the interplay between the large T-antigens and small t-antigens with host signaling pathways and the biological consequences of these interactions.
Collapse
Affiliation(s)
- Ugo Moens
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway.
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
206
|
Liu J, Han X, Zhu G, Liu S, Lu Q, Tang Z. Analysis of potential functional significance of microRNA‑3613‑3p in human umbilical vein endothelial cells affected by heat stress. Mol Med Rep 2019; 20:1846-1856. [PMID: 31257536 PMCID: PMC6625459 DOI: 10.3892/mmr.2019.10376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 04/24/2019] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of microRNA‑3613‑3p (miR‑3613‑3p) was previously reported in endothelial cells (ECs) during heat stress. The aim of the present study was to investigate the precise role of miR‑3613‑3p in heat stress. In the present study, potential gene targets of miR‑3613‑3p in heat‑treated ECs were assessed, and the potential effects of miR‑3613‑3p were determined using Gene Ontology enrichment analysis. Kyoto Encyclopedia of Genes and Genomes pathway analysis was used to identify signaling pathways that may be affected by miR‑3613‑3p in heat‑treated cells. Reverse transcription‑quantitative PCR, western blotting and annexin V‑FITC/propidium iodide staining were performed to detect miRNA expression, protein expression and apoptosis, respectively. Luciferase gene reporter assay was performed to evaluate the association between miR‑3613‑3p and mitogen‑activated protein kinase kinase kinase 2 (MAP3K2). Bioinformatics analysis revealed 865 potential gene targets for miR‑3613‑3p and a series of functions and pathways in heat‑treated ECs. 'Negative regulation of apoptotic process' was identified as a potential function of miR‑3613‑3p. In addition, functional analysis confirmed the downregulated expression levels of miR‑3613‑3p in ECs during heat stress, which was accompanied by an increase in apoptosis; restoration of miR‑3613‑3p expression inhibited apoptosis. MAP3K2 protein was demonstrated to be upregulated in heat‑treated ECs, and overexpression of miR‑3613‑3p reduced MAP3K2 expression levels. Additionally, MAP3K2 was targeted by miR‑3613‑3p. These results indicated that miR‑3613‑3p may have complicated roles in ECs under heat stress. miR‑3613‑3p may serve an important role in the apoptosis of heat‑treated ECs, and this effect may be partly achieved by targeting MAP3K2.
Collapse
Affiliation(s)
- Jie Liu
- Department of Emergency, Central Theater General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
- Department of Emergency, Hefei BOE Hospital Co., Ltd., Anhui, Hefei 230011, P.R. China
| | - Xuan Han
- Department of Emergency, Central Theater General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Guoguo Zhu
- Department of Emergency, Central Theater General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Shixin Liu
- Department of Emergency, Central Theater General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Qiping Lu
- Department of General Surgery, Central Theater General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Zhongzhi Tang
- Department of Emergency, Central Theater General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
207
|
Du L, Anderson A, Nguyen K, Ojeda SS, Ortiz-Rivera I, Nguyen TN, Zhang T, Kaoud TS, Gray NS, Dalby KN, Tsai KY. JNK2 Is Required for the Tumorigenic Properties of Melanoma Cells. ACS Chem Biol 2019; 14:1426-1435. [PMID: 31063355 DOI: 10.1021/acschembio.9b00083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Overexpression and activation of c-Jun N-terminal kinases (JNKs) have been observed in multiple cancer cell lines and tumor samples. Various JNK isoforms have been reported to promote lung and liver cancer, as well as keratinocyte transformation, suggesting an important role of JNK signaling in promoting tumor development. However, there are three JNK isoforms, and it is unclear how each individual isoform, especially the ubiquitously expressed JNK1 and JNK2, functions in melanoma. Our previous study found that C116S mutations in both JNK1 and JNK2 rendered them insensitive to the covalent pan-JNK inhibitor JNK-IN-8 while retaining kinase activity. To delineate the specific roles of JNK1 and JNK2 in melanoma cell proliferation and invasiveness, we expressed the wild type (WT) and C116S mutants in melanoma cell lines and used JNK-IN-8 to enable chemical-genetic dissection of JNK1 and JNK2 activity. We found that the JNK2C116S allele consistently enhanced colony proliferation and cell invasiveness in the presence of JNK-IN-8. When cells individually expressing WT or C116S JNK1/2 were subcutaneously implanted into immunodeficient mice, we again found that bypass of JNK-IN-8-mediated inhibition of JNK signaling by expression of JNK2C116S specifically resulted in enhanced tumor growth in vivo. In addition, we observed a high level of JNK pathway activation in some human BRAF inhibitor (BRAFi) resistant melanoma cell lines relative to their BRAFi sensitive isogenic counterparts. JNK-IN-8 significantly enhanced the response to dabrafenib in resistant cells overexpressing JNK1WT, JNK2WT, and JNK1C116S but had no effect on cells expressing JNK2C116S, suggesting that JNK2 signaling is also crucial for BRAFi resistance in a subset of melanomas. Collectively, our data show that JNK2 activity is specifically required for melanoma cell proliferation, invasiveness, and BRAFi resistance and that this activity is most important in the context of JNK1 suppression, thus providing a compelling rationale for the development of JNK2 selective inhibitors as a potential therapy for the treatment of melanoma.
Collapse
Affiliation(s)
- Lili Du
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, United States
| | - Anna Anderson
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, United States
| | - Kimberly Nguyen
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, United States
- Departments of Anatomic Pathology and Tumor Biology, Co-Director, Donald A. Adam Melanoma & Skin Cancer Center of Excellence, Moffitt Cancer Center, Tampa, Florida 33612, United States
| | - Sandra S. Ojeda
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, United States
| | - Ivannie Ortiz-Rivera
- Departments of Anatomic Pathology and Tumor Biology, Co-Director, Donald A. Adam Melanoma & Skin Cancer Center of Excellence, Moffitt Cancer Center, Tampa, Florida 33612, United States
| | - Tran Ngoc Nguyen
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, United States
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Tamer S. Kaoud
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Kevin N. Dalby
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kenneth Y. Tsai
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, United States
- Departments of Anatomic Pathology and Tumor Biology, Co-Director, Donald A. Adam Melanoma & Skin Cancer Center of Excellence, Moffitt Cancer Center, Tampa, Florida 33612, United States
| |
Collapse
|
208
|
Batlle R, Andrés E, Gonzalez L, Llonch E, Igea A, Gutierrez-Prat N, Berenguer-Llergo A, Nebreda AR. Regulation of tumor angiogenesis and mesenchymal-endothelial transition by p38α through TGF-β and JNK signaling. Nat Commun 2019; 10:3071. [PMID: 31296856 PMCID: PMC6624205 DOI: 10.1038/s41467-019-10946-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/12/2019] [Indexed: 12/17/2022] Open
Abstract
The formation of new blood vessels is essential for normal development, tissue repair and tumor growth. Here we show that inhibition of the kinase p38α enhances angiogenesis in human and mouse colon tumors. Mesenchymal cells can contribute to tumor angiogenesis by regulating proliferation and migration of endothelial cells. We show that p38α negatively regulates an angiogenic program in mesenchymal stem/stromal cells (MSCs), multipotent progenitors found in perivascular locations. This program includes the acquisition of an endothelial phenotype by MSCs mediated by both TGF-β and JNK, and negatively regulated by p38α. Abrogation of p38α in mesenchymal cells increases tumorigenesis, which correlates with enhanced angiogenesis. Using genetic models, we show that p38α regulates the acquisition of an endothelial-like phenotype by mesenchymal cells in colon tumors and damage tissue. Taken together, our results indicate that p38α in mesenchymal cells restrains a TGF-β-induced angiogenesis program including their ability to transdifferentiate into endothelial cells. Mesenchymal cells contribute to tumor angiogenesis by regulating proliferation and migration of endothelial cells. Here, the authors show that mesenchymal stem cells also have the ability to acquire an endothelial phenotype upon TGF-β stimulation via the downstream kinase JNK, and that p38α negatively regulates this process.
Collapse
Affiliation(s)
- Raquel Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Eva Andrés
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Lorena Gonzalez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Elisabet Llonch
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Ana Igea
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Núria Gutierrez-Prat
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Antoni Berenguer-Llergo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain. .,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
209
|
Escitalopram Ameliorates Cognitive Impairment in D-Galactose-Injected Ovariectomized Rats: Modulation of JNK, GSK-3β, and ERK Signalling Pathways. Sci Rep 2019; 9:10056. [PMID: 31296935 PMCID: PMC6624366 DOI: 10.1038/s41598-019-46558-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/27/2019] [Indexed: 12/29/2022] Open
Abstract
Though selective serotonin reuptake inhibitors (SSRIs) have been found to increase cognitive performance in some studies on patients and animal models of Alzheimer's disease (AD), other studies have reported contradictory results, and the mechanism of action has not been fully described. This study aimed to examine the effect of escitalopram, an SSRI, in an experimental model of AD and to determine the involved intracellular signalling pathways. Ovariectomized rats were administered D-galactose (150 mg/kg/day, i.p) over ten weeks to induce AD. Treatment with escitalopram (10 mg/kg/day, p.o) for four weeks, starting from the 7th week of D-galactose injection, enhanced memory performance and attenuated associated histopathological changes. Escitalopram reduced hippocampal amyloid β 42, β-secretase, and p-tau, while increasing α-secretase levels. Furthermore, it decreased tumor necrosis factor-α, nuclear factor-kappa B p65, and NADPH oxidase, while enhancing brain-derived neurotrophic factor, phospho-cAMP response element binding protein, and synaptophysin levels. Moreover, escitalopram diminished the protein expression of the phosphorylated forms of c-Jun N-terminal kinase (JNK)/c-Jun, while increasing those of phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), glycogen synthase kinase-3β (GSK-3β), extracellular signal-regulated kinase (ERK) and its upstream kinases MEK and Raf-1. In conclusion, escitalopram ameliorated D-galactose/ovariectomy-induced AD-like features through modulation of PI3K/Akt/GSK-3β, Raf-1/MEK/ERK, and JNK/c-Jun pathways.
Collapse
|
210
|
Lin H, Lu P, Zhou M, Wu F, Weng L, Meng K, Yang D, Li S, Jiang C, Tian H. Purification of recombinant human fibroblast growth factor 13 in E. coli and its molecular mechanism of mitogenesis. Appl Microbiol Biotechnol 2019; 103:7017-7027. [PMID: 31289905 DOI: 10.1007/s00253-019-09973-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/27/2019] [Accepted: 06/06/2019] [Indexed: 12/19/2022]
Abstract
Fibroblast growth factor (FGF) 13, a member of the FGF11 subfamily, is a kind of intracrine protein similar to other family members including FGF11, FGF12, and FGF14. Unlike classical FGF, FGF13 exerts its bioactivities independent of fibroblast growth factor receptors (FGFRs). However, the effect of exogenous administration of FGF13 still remains further investigated. In the present study, we established an Escherichia coli expression system for the large-scale production of FGF13 and then obtained two isoform proteins including recombinant human FGF13A (rhFGF13A) and rhFGF13B with a purity greater than 90% by column chromatography, respectively. Otherwise, soluble analysis indicated that both rhFGF13A and rhFGF13B expressed in E. coli BL21 (DE3) pLysS were soluble. Furthermore, cellular-based experiments demonstrated that rhFGF13A, rather than rhFGF13B, could promote the proliferation of NIH3T3 cells in the presence of heparin. Mechanistically, the mitogenic effect of FGF13 was mediated by activation of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), but not p38. Moreover, blockage of FGFRs also significantly attenuated the mitogenic effects of rhFGF13A, implying that FGFRs are still related to FGF13. Thus, our research shows that exogenous FGF13 can act as secreted FGF to participate in cell signal transmission and heparin is still required as an ancillary cofactor for the mitogenic effects of FGF13, which may help people to discover more potential functions of FGF13 in cell life activities.
Collapse
Affiliation(s)
- Haipeng Lin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Panyu Lu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Mi Zhou
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fenfang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Lei Weng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Kuikui Meng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Dan Yang
- Biomedicine Collaborative Innovation Center, Wenzhou University, Wenzhou, 325035, Zhejiang, China
| | - Shijun Li
- Biomedicine Collaborative Innovation Center, Wenzhou University, Wenzhou, 325035, Zhejiang, China
| | - Chao Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China. .,Biomedicine Collaborative Innovation Center, Wenzhou University, Wenzhou, 325035, Zhejiang, China.
| | - Haishan Tian
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
211
|
Wang H, Abel GM, Storm DR, Xia Z. Cadmium Exposure Impairs Adult Hippocampal Neurogenesis. Toxicol Sci 2019; 171:501-514. [PMID: 31271426 DOI: 10.1093/toxsci/kfz152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/07/2019] [Accepted: 06/29/2019] [Indexed: 01/30/2023] Open
Abstract
Cadmium (Cd) is an environmental pollutant of considerable interest throughout the world and potentially a neurotoxicant. Our recent data indicate that Cd exposure induces impairment of hippocampus-dependent learning and memory in mice. However, the underlying mechanisms for this defect are not known. The goal of this study was to determine if Cd inhibits adult neurogenesis and to identify underlying signaling pathways responsible for this impairment. Adult hippocampal neurogenesis is a process in which adult neural progenitor/stem cells (aNPCs) in the subgranular zone (SGZ) of the dentate gyrus (DG) generate functional new neurons in the hippocampus which contributes to hippocampus-dependent learning and memory. However, studies concerning the effects of neurotoxicants on adult hippocampal neurogenesis and the underlying signaling mechanisms are limited. Here, we report that Cd significantly induces apoptosis, inhibits proliferation, and impairs neuronal differentiation in primary cultured aNPCs derived from the SGZ. In addition, the c-jun NH2-terminal kinase (JNK) and p38 mitogen-activated protein (MAP) kinase signaling pathways are activated by Cd and contribute to its toxicity. Furthermore, we exposed 8-week-old male C57BL/6 mice to Cd through drinking water for 13 weeks to assess the effects of Cd on adult hippocampal neurogenesis in vivo. Cd treatment reduced the number of 5-week old adult-born cells in the DG and impaired the differentiation of adult-born hippocampal neurons. These results suggest that Cd exposure impairs adult hippocampal neurogenesis both in vitro and in vivo. This may contribute to Cd-mediated inhibition of hippocampus-dependent learning and memory.
Collapse
Affiliation(s)
- Hao Wang
- Toxicology Program, Department of Environmental and Occupational Health Sciences
| | - Glen M Abel
- Toxicology Program, Department of Environmental and Occupational Health Sciences
| | - Daniel R Storm
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Zhengui Xia
- Toxicology Program, Department of Environmental and Occupational Health Sciences
| |
Collapse
|
212
|
Li P, Ma Z, Yu Y, Hu X, Zhou Y, Song H. FER promotes cell migration via regulating JNK activity. Cell Prolif 2019; 52:e12656. [PMID: 31264309 PMCID: PMC6797522 DOI: 10.1111/cpr.12656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/11/2022] Open
Abstract
Objectives Cell migration has a key role in cancer metastasis, which contributes to drug resistance and tumour recurrence. Better understanding of the mechanisms involved in this process will potentially reveal new drug targets for cancer therapy. Fer is a non‐receptor protein tyrosine kinase aberrantly expressed in various human cancers, whereas its role in tumour progression remains elusive. Materials and Methods Transgenic flies and epigenetic analysis were employed to investigate the role of Drosophila Fer (FER) in cell migration and underlying mechanisms. Co‐immunoprecipitation assay was used to monitor the interaction between FER and Drosophila JNK (Bsk). The conservation of Fer in regulating JNK signalling was explored in mammalian cancer and non‐cancer cells. Results Overexpression of FER triggered cell migration and activated JNK signalling in the Drosophila wing disc. Upregulation and downregulation in the basal activity of Bsk exacerbated and eliminated FER‐mediated migration, respectively. In addition, loss of FER blocked signal transduction of the JNK pathway. Specifically, FER interacted with and promoted the activity of Bsk, which required both the kinase domain and the C‐terminal of Bsk. Lastly, Fer regulated JNK activities in mammalian cells. Conclusions Our study reveals FER as a positive regulator of JNK‐mediated cell migration and suggests its potential role as a therapeutic target for cancer metastasis.
Collapse
Affiliation(s)
- Ping Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhiwei Ma
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yun Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xingjie Hu
- School of Public Health, Guangzhou Medical University, Guangdong, China
| | - Yanfeng Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyun Song
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
213
|
Lu M, Wang Y, Zhan X. The MAPK Pathway-Based Drug Therapeutic Targets in Pituitary Adenomas. Front Endocrinol (Lausanne) 2019; 10:330. [PMID: 31231308 PMCID: PMC6558377 DOI: 10.3389/fendo.2019.00330] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) include ERK, p38, and JNK MAPK subfamilies, which are crucial regulators of cellular physiology, cell pathology, and many diseases including cancers. For the MAPK signaling system in pituitary adenomas (PAs), the activation of ERK signaling is generally thought to promote cell proliferation and growth; whereas the activations of p38 and JNK signaling are generally thought to promote cell apoptosis. The role of MAPK in treatment of PAs is demonstrated through the effects of currently used medications such as somatostatin analogs such as SOM230 and OCT, dopamine agonists such as cabergoline and bromocriptine, and retinoic acid which inhibit the MAPK pathway. Further, there are potential novel therapies based on putative molecular targets of the MAPK pathway, including 18beta-glycyrrhetinic acid (GA), dopamine-somatostatin chimeric compound (BIM-23A760), ursolic acid (UA), fulvestrant, Raf kinase inhibitory protein (RKIP), epidermal growth factor pathway substrate number 8 (Eps8), transmembrane protein with EGF-like and two follistatin-like domains (TMEFF2), cold inducible RNA-binding protein (CIRP), miR-16, and mammaliansterile-20-like kinase (MST4). The combined use of ERK inhibitor (e.g., SOM230, OCT, or dopamine) plus p38 activator (e.g., cabergoline, bromocriptine, and fulvestrant) and/or JNK activator (e.g., UA), or the development of single drug (e.g., BIM-23A760) to target both ERK and p38 or JNK pathways, might produce better anti-tumor effects on PAs. This article reviews the advances in understanding the role of MAPK signaling in pituitary tumorigenesis, and the MAPK pathway-based potential therapeutic drugs for PAs.
Collapse
Affiliation(s)
- Miaolong Lu
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Ya Wang
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
214
|
Oscillating flow promotes inflammation through the TLR2–TAK1–IKK2 signalling pathway in human umbilical vein endothelial cell (HUVECs). Life Sci 2019; 224:212-221. [DOI: 10.1016/j.lfs.2019.03.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 03/10/2019] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
|
215
|
Tsai CR, Wang Y, Galko MJ. Crawling wounded: molecular genetic insights into wound healing from Drosophila larvae. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2019; 62:479-489. [PMID: 29938760 PMCID: PMC6352908 DOI: 10.1387/ijdb.180085mg] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
For animals, injury is inevitable. Because of this, organisms possess efficient wound healing mechanisms that can repair damaged tissues. However, the molecular and genetic mechanisms by which epidermal repair is accomplished remain poorly defined. Drosophila has become a valuable model to study epidermal wound healing because of the comprehensive genetic toolkit available in this organism and the similarities of wound healing processes between Drosophila and vertebrates. Other reviews in this Special Issue cover wound healing assays and pathways in Drosophila embryos, pupae and adults, as well as regenerative processes that occur in tissues such as imaginal discs and the gut. In this review, we will focus on the molecular/genetic control of wound-induced cellular processes such as inflammation, cell migration and epithelial cell-cell fusion in Drosophila larvae. We will give a brief overview of the three wounding assays, pinch, puncture, and laser ablation, and the cellular responses that ensue following wounding. We will highlight the actin regulators, signaling pathways and transcriptional mediators found so far to be involved in larval epidermal wound closure and what is known about how they act. We will also discuss wound-induced epidermal cell-cell fusion and possible directions for future research in this exciting system.
Collapse
Affiliation(s)
- Chang-Ru Tsai
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | | |
Collapse
|
216
|
Liu Q, Jiang L, Wang W, Jiang T. BTF3 Silencing Inhibits the Proliferation of Osteosarcoma Cells. J Cancer 2019; 10:1855-1861. [PMID: 31205542 PMCID: PMC6547992 DOI: 10.7150/jca.28476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 02/22/2019] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is one of the bone malignancy cancers with poor prognosis in the early stages worldwide. Basic transcription factor 3 (BTF3) is associated with the development of several types of cancer. The present study aimed to evaluate the role of BTF3 in OS. Silencing of BTF3 was achieved by using stable lentivirus transfection of siRNA targeting BTF3 in the human OS cell line Saos-2. Cell viability and colony-forming ability were measured using methyl-thaizolyl-tetrazolium (MTT) and colony formation assays, respectively. Propidium iodide staining and flow cytometry was used to detect the progression of the cell cycle. To evaluate the possible intracellular signaling molecules involved, a PathScan Intracellular Signaling Array Kit was utilized. Lentivirus-BTF3-shRNA (LV-BTF3-shRNA) suppressed expression of BTF3 in Saos-2 cells (inhibition ratio: 89.8%), which significantly inhibited cell proliferation (48.5%), colony formation and enhanced apoptosis to 48.2% compared to 4.5% with lentivirus control shRNA (N-shRNA). Additionally, BTF3 silencing enhanced the percentage of Saos-2 cells in S and G2/M phases, but significantly reduced cells in the G0/M phase (all P < 0.01). The proteins activated by BTF3 included STAT3, S6 ribosomal protein, HSP27 and SAPK/JNK2, all of which were inhibited by BTF3 silencing, whereas SAPK/JNK1 was upregulated by BTF3 silencing. In the present study, we explored the crucial role of BTF3 in promoting OS cell proliferation as well as laying the foundations for further research to investigate the clinical potential of lentivirus-mediated delivery of BTF3 interruption therapy for the treatment of OS.
Collapse
Affiliation(s)
- Qi Liu
- Department of General Surgery, People's Hospital of Hunan Province, First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Lin Jiang
- Department of Orthopedic Surgery, Traumatology and Orthopedics Hospital of Liuyang, Liuyang 410300, Hunan Province, China
| | - Wanchun Wang
- Department of Orthopedic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410005, Hunan Province, China
| | - Tao Jiang
- Department of Orthopedic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410005, Hunan Province, China
| |
Collapse
|
217
|
Park E, Gim J, Kim DK, Kim CS, Chun HS. Protective Effects of Alpha-Lipoic Acid on Glutamate-Induced Cytotoxicity in C6 Glioma Cells. Biol Pharm Bull 2019; 42:94-102. [PMID: 30606992 DOI: 10.1248/bpb.b18-00603] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glutamate-mediated cytotoxicity has been implicated in the pathogenesis of neurological diseases, including Parkinson's disease, Alzheimer's disease, and stroke. In this study, we investigated the protective effects of alpha-lipoic acid (ALA), a naturally occurring thiol antioxidant, on glutamate-induced cytotoxicity in cultured C6 astroglial cells. Exposure to high-dose glutamate (10 mM) caused oxidative stress and mitochondrial dysfunction through the elevation of reactive oxygen species, depletion of glutathione, and loss of the mitochondrial membrane potential (ΔΨm). Pretreatment with ALA (200 µM), however, significantly inhibited the glutamate-induced oxidative stress and mitochondrial dysfunction. ALA pretreatment dose-dependently suppressed glutamate-induced apoptotic events including altered nuclear morphology and activation of caspase-3. In addition, ALA significantly attenuated glutamate-induced endoplasmic reticulum (ER) stress markers; namely, glucose-regulated protein 78 (GRP78), activating transcription factor 6 (ATF6), protein kinase regulated by RNA (PKR)-like ER-associated kinase (PERK), eukaryotic translation initiation factor 2 alpha (eIF2α), inositol-requiring enzyme 1 (IRE1), CCAAT/enhancer binding protein homologous protein (CHOP), and caspase-12. We confirmed that CHOP and caspase-12 are key mediators of glutamate-induced ER stress. Furthermore, exposure of the cells to a caspase-12-specific inhibitor and CHOP small interfering RNAs (siRNAs) led to restoration of the ΔΨm that was damaged by glutamate treatment. These results suggest that ALA can effectively suppress oxidative stress, mitochondrial dysfunction, and ER stress in astroglial cells.
Collapse
Affiliation(s)
- Euteum Park
- Department of Biomedical Science, Chosun University
| | - Jungsoo Gim
- Department of Biomedical Science, Chosun University
| | - Do Kyung Kim
- Department of Oral Physiology, College of Dentistry, Chosun University
| | - Chun-Sung Kim
- Department of Oral Biochemistry, College of Dentistry, Chosun University
| | | |
Collapse
|
218
|
Wang Z, Shen J, Sun W, Zhang T, Zuo D, Wang H, Wang G, Xu J, Yin F, Mao M, Zhou Z, Hua Y, Cai Z. Antitumor activity of Raddeanin A is mediated by Jun amino-terminal kinase activation and signal transducer and activator of transcription 3 inhibition in human osteosarcoma. Cancer Sci 2019; 110:1746-1759. [PMID: 30907478 PMCID: PMC6500987 DOI: 10.1111/cas.14008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 01/01/2023] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor. Raddeanin A (RA) is an active oleanane‐type triterpenoid saponin extracted from the traditional Chinese herb Anemone raddeana Regel that exerts antitumor activity against several cancer types. However, the effect of RA on osteosarcoma remains unclear. In the present study, we showed that RA inhibited proliferation and induced apoptosis of osteosarcoma cells in a dose‐ and time‐dependent way in vitro and in vivo. RA treatment resulted in excessive reactive oxygen species (ROS) generation and JNK and ERK1/2 activation. Apoptosis induction was evaluated by the activation of caspase‐3, caspase‐8, and caspase‐9 and poly‐ADP ribose polymerase (PARP) cleavage. RA‐induced cell death was significantly restored by the ROS scavenger glutathione (GSH), the pharmacological inhibitor of JNK SP600125, or specific JNK knockdown by shRNA. Additionally, signal transducer and activator of transcription 3 (STAT3) activation was suppressed by RA in human osteosarcoma, and this suppression was restored by GSH, SP600125, and JNK‐shRNA. Further investigation showed that STAT3 phosphorylation was increased after JNK knockdown. In a tibial xenograft tumor model, RA induced osteosarcoma apoptosis and notably inhibited tumor growth. Taken together, our results show that RA suppresses proliferation and induces apoptosis by modulating the JNK/c‐Jun and STAT3 signaling pathways in human osteosarcoma. Therefore, RA may be a promising candidate antitumor drug for osteosarcoma intervention.
Collapse
Affiliation(s)
- Zhuoying Wang
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiakang Shen
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Wei Sun
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Zhang
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongqing Zuo
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongsheng Wang
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gangyang Wang
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xu
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Yin
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Mao
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zifei Zhou
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingqi Hua
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengdong Cai
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
219
|
Sun X, Yan P, Zou C, Wong YK, Shu Y, Lee YM, Zhang C, Yang ND, Wang J, Zhang J. Targeting autophagy enhances the anticancer effect of artemisinin and its derivatives. Med Res Rev 2019; 39:2172-2193. [PMID: 30972803 DOI: 10.1002/med.21580] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 12/12/2022]
Abstract
Artemisinin and its derivatives, with their outstanding clinical efficacy and safety, represent the most effective and impactful antimalarial drugs. Apart from its antimalarial effect, artemisinin has also been shown to exhibit selective anticancer properties against multiple cancer types both in vitro and in vivo. Specifically, our previous studies highlighted the therapeutic effects of artemisinin on autophagy regulation. Autophagy is a well-conserved degradative process that recycles cytoplasmic contents and organelles in lysosomes to maintain cellular homeostasis. The deregulation of autophagy is often observed in cancer cells, where it contributes to tumor adaptation to nutrient-deficient tumor microenvironments. This review discusses recent advances in the anticancer properties of artemisinin and its derivatives via their regulation of autophagy, mitophagy, and ferritinophagy. In particular, we will discuss the mechanisms of artemisinin activation in cancer and novel findings regarding the role of artemisinin in regulating autophagy, which involves changes in multiple signaling pathways. More importantly, with increasing failure rates and the high cost of the development of novel anticancer drugs, the strategy of repurposing traditional therapeutic Chinese medicinal agents such as artemisinin to treat cancer provides a more attractive alternative. We believe that the topics covered here will be important in demonstrating the potential of artemisinin and its derivatives as safe and potent anticancer agents.
Collapse
Affiliation(s)
- Xin Sun
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Peiyi Yan
- Department of Clinical Laboratory, Shanghai Putuo District People's Hospital, Shanghai, China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University, Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, Shenzhen, China
| | - Yin-Kwan Wong
- Department of Pharmacology, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuhan Shu
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Yew Mun Lee
- Department of Pharmacology, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chongjing Zhang
- Institute of Material Medical, Peking Union Medical College, Beijing, China
| | - Nai-Di Yang
- Department of Pharmacology, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigang Wang
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University, Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, Shenzhen, China.,Department of Pharmacology, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Cardio-Cerebrovascular Disease Prevention & Therapy, Gannan Medical University, Ganzhou, China
| | - Jianbin Zhang
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
220
|
Sun Y, Zhang D, Li C, Huang J, Li W, Qiu Y, Mao A, Zhou M, Xue L. Lic regulates JNK-mediated cell death in Drosophila. Cell Prolif 2019; 52:e12593. [PMID: 30847993 PMCID: PMC6536442 DOI: 10.1111/cpr.12593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 01/01/2023] Open
Abstract
Objectives The evolutionary conserved JNK pathway plays crucial role in cell death, yet factors that modulate this signalling have not been fully disclosed. In this study, we aim to identify additional factors that regulate JNK signalling in cell death, and characterize the underlying mechanisms. Materials and Methods Drosophila were raised on standard media, and cross was carried out at 25°C. The Gal4/UAS system was used to express proteins or RNAi in a specific temporal and spatial pattern. Gene expression was revealed by GFP fluorescence, X‐gal staining or immunostaining of 3rd instar larval eye and wing discs. Cell death was visualized by acridine orange (AO) staining. Images of fly eyes and wings were taken by OLYMPUS microscopes. Results We found that licorne (lic) encoding the Drosophila MKK3 is an essential regulator of JNK‐mediated cell death. Firstly, loss of lic suppressed ectopic Egr‐triggered JNK activation and cell death in eye and wing development. Secondary, lic is necessary for loss‐of‐cell polarity‐induced, physiological JNK‐dependent cell death in wing development. Thirdly, Lic overexpression is sufficient to initiate JNK‐mediated cell death in developing eyes and wings. Furthermore, ectopic Lic activates JNK signalling by promoting JNK phosphorylation. Finally, genetic epistatic analysis confirmed that Lic acts in parallel with Hep in the Egr‐JNK pathway. Conclusions This study not only identified Lic as a novel component of the JNK signalling, but also disclosed the crucial roles and mechanism of Lic in cell death.
Collapse
Affiliation(s)
- Yihao Sun
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Di Zhang
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Chenglin Li
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Jiuhong Huang
- International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing, China
| | - Wenzhe Li
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yu Qiu
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Aiwu Mao
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingcheng Zhou
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Lei Xue
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| |
Collapse
|
221
|
Mirzoyan Z, Sollazzo M, Allocca M, Valenza AM, Grifoni D, Bellosta P. Drosophila melanogaster: A Model Organism to Study Cancer. Front Genet 2019; 10:51. [PMID: 30881374 PMCID: PMC6405444 DOI: 10.3389/fgene.2019.00051] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/21/2019] [Indexed: 12/26/2022] Open
Abstract
Cancer is a multistep disease driven by the activation of specific oncogenic pathways concomitantly with the loss of function of tumor suppressor genes that act as sentinels to control physiological growth. The conservation of most of these signaling pathways in Drosophila, and the ability to easily manipulate them genetically, has made the fruit fly a useful model organism to study cancer biology. In this review we outline the basic mechanisms and signaling pathways conserved between humans and flies responsible of inducing uncontrolled growth and cancer development. Second, we describe classic and novel Drosophila models used to study different cancers, with the objective to discuss their strengths and limitations on their use to identify signals driving growth cell autonomously and within organs, drug discovery and for therapeutic approaches.
Collapse
Affiliation(s)
- Zhasmine Mirzoyan
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Manuela Sollazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Mariateresa Allocca
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | | | - Daniela Grifoni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Department of Biosciences, University of Milan, Milan, Italy.,Department of Medicine, NYU Langone Medical Center, New York, NY, United States
| |
Collapse
|
222
|
Roskoski R. Targeting ERK1/2 protein-serine/threonine kinases in human cancers. Pharmacol Res 2019; 142:151-168. [PMID: 30794926 DOI: 10.1016/j.phrs.2019.01.039] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 12/17/2022]
Abstract
ERK1 and ERK2 are key protein kinases that contribute to the Ras-Raf-MEK-ERK MAP kinase signalling module. This pathway participates in the control of numerous processes including apoptosis, cell proliferation, the immune response, nervous system function, and RNA synthesis and processing. MEK1/2 activate human ERK1/2 by first catalyzing the phosphorylation of Y204/187 and then T202/185, both residues of which occur within the activation segment. The phosphorylation of both residues is required for enzyme activation. The only Raf substrates are MEK1/2 and the only MEK1/2 substrates are ERK1/2. In contrast, ERK1/2 catalyze the phosphorylation of many cytoplasmic and nuclear substrates including transcription factors and regulatory molecules. The linear MAP kinase pathway branches extensively at the ERK1/2 node. ERK1/2 are proline-directed kinases that preferentially catalyze the phosphorylation of substrates containing a PxS/TP sequence. The dephosphorylation and inactivation of ERK1/2 is catalyzed by dual specificity phosphatases, protein-tyrosine specific phosphatases, and protein-serine/threonine phosphatases. The combined functions of kinases and phosphatases make the overall process reversible. To provide an idea of the complexities involved in these reactions, somatic cell cycle progression involves the strict timing of more than 32,000 phosphorylation and dephosphorylation events as determined by mass spectrometry. The MAP kinase cascade is perhaps the most important oncogenic driver of human cancers and the blockade of this signalling module by targeted inhibitors is an important anti-tumor strategy. Although numerous cancers are driven by MAP kinase pathway activation, thus far the only orally effective approved drugs that target this signaling module are used for the treatment of BRAF-mutant melanomas. The best treatments include the combination of B-Raf and MEK inhibitors (dabrafenib and trametinib, encorafenib and binimetinib, vemurafenib and cobimetanib). However, resistance to these antagonists occurs within one year and additional treatment options are necessary. Owing to the large variety of malignancies that are driven by dysregulation of the MAP kinase pathway, additional tumor types should be amenable to MAP kinase pathway inhibitor therapy. In addition to new B-Raf and MEK inhibitors, the addition of ERK inhibitors should prove helpful. Ulixertinib, MK-8353, and GDC-0994 are orally effective, potent, and specific inhibitors of ERK1/2 that are in early clinical trials for the treatment of various advanced/metastatic solid tumors. These agents are effective against cell lines that are resistant to B-Raf and MEK1/2 inhibitor therapy. Although MK-8353 does not directly inhibit MEK1/2, it decreases the phosphorylation of ERK1/2 as well as the phosphorylation of RSK, an ERK1/2 substrate. The decrease in RSK phosphorylation appears to be a result of ERK inhibition and the decrease in ERK1/2 phosphorylation is related to the inability of MEK to catalyze the phosphorylation of the ERK-MK-8353 complex; these decreases characterize the ERK dual mechanism inhibition paradigm. Additional work will be required to determine whether ERK inhibitors will be successful in the clinic and are able to forestall the development of drug resistance of the MAP kinase pathway.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 116, Box 19, Horse Shoe, NC, 28742-8814, United States.
| |
Collapse
|
223
|
Giampieri F, Afrin S, Forbes-Hernandez TY, Gasparrini M, Cianciosi D, Reboredo-Rodriguez P, Varela-Lopez A, Quiles JL, Battino M. Autophagy in Human Health and Disease: Novel Therapeutic Opportunities. Antioxid Redox Signal 2019; 30:577-634. [PMID: 29943652 DOI: 10.1089/ars.2017.7234] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE In eukaryotes, autophagy represents a highly evolutionary conserved process, through which macromolecules and cytoplasmic material are degraded into lysosomes and recycled for biosynthetic or energetic purposes. Dysfunction of the autophagic process has been associated with the onset and development of many human chronic pathologies, such as cardiovascular, metabolic, and neurodegenerative diseases as well as cancer. Recent Advances: Currently, comprehensive research is being carried out to discover new therapeutic agents that are able to modulate the autophagic process in vivo. Recent evidence has shown that a large number of natural bioactive compounds are involved in the regulation of autophagy by modulating several transcriptional factors and signaling pathways. CRITICAL ISSUES Critical issues that deserve particular attention are the inadequate understanding of the complex role of autophagy in disease pathogenesis, the limited availability of therapeutic drugs, and the lack of clinical trials. In this context, the effects that natural bioactive compounds exert on autophagic modulation should be clearly highlighted, since they depend on the type and stage of the pathological conditions of diseases. FUTURE DIRECTIONS Research efforts should now focus on understanding the survival-supporting and death-promoting roles of autophagy, how natural compounds interact exactly with the autophagic targets so as to induce or inhibit autophagy and on the evaluation of their pharmacological effects in a more in-depth and mechanistic way. In addition, clinical studies on autophagy-inducing natural products are strongly encouraged, also to highlight some fundamental aspects, such as the dose, the duration, and the possible synergistic action of these compounds with conventional therapy.
Collapse
Affiliation(s)
- Francesca Giampieri
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Sadia Afrin
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Tamara Y Forbes-Hernandez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,2 Area de Nutricion y Salud, Universidad Internacional Iberoamericana , Campeche, Mexico
| | - Massimiliano Gasparrini
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Danila Cianciosi
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Patricia Reboredo-Rodriguez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,3 Departamento de Quimica Analıtica y Alimentaria, Grupo de Nutricion y Bromatologıa, Universidade Vigo , Ourense, Spain
| | - Alfonso Varela-Lopez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Jose L Quiles
- 4 Department of Physiology, Institute of Nutrition and Food Technology "Jose Mataix," Biomedical Research Centre, University of Granada , Granada, Spain
| | - Maurizio Battino
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,5 Centre for Nutrition and Health, Universidad Europea del Atlantico (UEA) , Santander, Spain
| |
Collapse
|
224
|
Xu B, Lian S, Guo JR, Wang JF, Zhang LP, Li SZ, Yang HM. Activation of the MAPK signaling pathway induces upregulation of pro-apoptotic proteins in the hippocampi of cold stressed adolescent mice. Neurosci Lett 2019; 699:97-102. [PMID: 30711527 DOI: 10.1016/j.neulet.2018.12.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 11/30/2022]
Abstract
Stress induces many non-specific responses in the hippocampus, especially during adolescence. Low environmental temperature is known to induce stress, but its influence on the hippocampus, especially in adolescent mice is not clear. We compared apoptotic-related protein levels and MAPK signaling pathway activation in hippocampal neurons of adolescent mice under low temperature conditions (4 °C for 12 h) with western blotting and immunohistochemistry. Western bolt results demonstrated that the levels of phospho-JNK, phospho-p38, and cleaved-caspase 3 significantly increased, while the ratio of Bcl-XL/Bax decreased, in the cold stress group. The results of immunohistochemistry (IHC) and Nissl staining demonstrated that the protein optical density of caspase 3 increased and Nissl bodies decreased in the cold stress group compared with controls. Thus, we conclude that cold exposure initiates activation of the MAPK signaling pathway and subsequently induces the upregulation of pro-apoptotic proteins in the hippocampi of adolescent mice. Overall our study reveals the relationship between cold stress and apoptosis in adolescent mice.
Collapse
Affiliation(s)
- Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Jing-Ru Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Jian-Fa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Li-Ping Zhang
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Shi-Ze Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China.
| | - Huan-Min Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China.
| |
Collapse
|
225
|
Ramachandran PV, Savini M, Folick AK, Hu K, Masand R, Graham BH, Wang MC. Lysosomal Signaling Promotes Longevity by Adjusting Mitochondrial Activity. Dev Cell 2019; 48:685-696.e5. [PMID: 30713071 DOI: 10.1016/j.devcel.2018.12.022] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 09/09/2018] [Accepted: 12/27/2018] [Indexed: 12/24/2022]
Abstract
Lysosomes and mitochondria are both crucial cellular organelles for metabolic homeostasis and organism health. However, mechanisms linking their metabolic activities to promote organism longevity remain poorly understood. We discovered that the induction of specific lysosomal signaling mediated by a LIPL-4 lysosomal acid lipase and its lipid chaperone LBP-8 increases mitochondrial ß-oxidation to reduce lipid storage and promote longevity in Caenorhabditis elegans. We further discovered that increased mitochondrial ß-oxidation reduces mitochondrial electron transport chain complex II activity, contributing to the induction of reactive oxygen species in mitochondria (mtROS) and the longevity effect conferred by LIPL-4-LBP-8 signaling. Moreover, by activating the JUN-1 transcription factor downstream of mtROS, the LIPL-4-LBP-8 signaling pathway induces antioxidant targets and oxidative stress tolerance. Together, these results reveal regulatory mechanisms by which lysosomal signaling triggers adjustments in mitochondrial activity and suggest the significance of these metabolic adjustments for improving metabolic fitness, redox homeostasis, and longevity.
Collapse
Affiliation(s)
- Prasanna V Ramachandran
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Marzia Savini
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrew K Folick
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kuang Hu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruchi Masand
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brett H Graham
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng C Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
226
|
Dutta D, Mutsuddi M, Mukherjee A. Synergistic interaction of Deltex and Hrp48 leads to JNK activation. Cell Biol Int 2019; 43:350-357. [PMID: 30597717 DOI: 10.1002/cbin.11089] [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] [Received: 09/21/2018] [Accepted: 12/23/2018] [Indexed: 01/10/2023]
Abstract
The communication among the cells plays a seminal role in metazoan development by coordinating multiple cellular processes that, in turn, helps in the maintenance of biological homeostasis. Our previous study demonstrated that Dx and Hrp48 together downregulate Notch signaling and induce cell death in Drosophila. To understand the signaling events behind the Dx and Hrp48-induced cell death in a greater detail, we performed a set of genetic experiments followed by immunocytochemical analyses. Our data revealed that Dx along with Hrp48 induced JNK activation and consequently cell death in the eye tissue. Additionally, using genetic and molecular approaches, we identified the domain of Dx protein responsible for its synergistic activity with Hrp48. Altogether, our analyses suggest that coexpression of Dx and Hrp48 activates JNK pathway to induce cell death in eye disc of Drosophila melanogaster.
Collapse
Affiliation(s)
- Debdeep Dutta
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| |
Collapse
|
227
|
Wingless modulates activator protein-1-mediated tumor invasion. Oncogene 2019; 38:3871-3885. [PMID: 30683884 DOI: 10.1038/s41388-018-0629-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022]
Abstract
Metastasis begins with a subset of local tumor cells acquiring the potential to invade into surrounding tissues, and remains to be a major obstacle for cancer treatments. More than 90% of cancer patients died from tumor metastasis, instead of primary tumor growth. The canonical Wnt/β-catenin pathway plays essential roles in promoting tumor formation, yet its function in regulating tumor metastasis and the underlying mechanisms remain controversial. Here we employed well-established Drosophila tumor models to investigate the regulating mechanism of Wingless (Wg) pathway in tumor invasion. Our results showed that Wg signaling is necessary and sufficient for cell polarity disruption-induced cell migration and molecular changes reminiscent of epithelial-mesenchymal transition (EMT). Moreover, reducing Wg signaling suppressed lgl-/-/RasV12-induced tumor invasion, and cooperation between Arm and RasV12 is sufficient to induce tumor invasion. Mechanistically, we found that cell polarity disruption activates JNK signaling, which in turn upregulate wg expression through transcription factor activator protein-1 (AP-1). We identified a consensus AP-1 binding site located in the 2nd intron of wg, and confirmed that it is essential for AP-1 induced wg transcription both in vitro and in vivo. Lastly, we confirmed that the transcriptional activation of WNT by AP-1 is conserved in human cancer cells. These evidences reveal a positive role of Wnt/β-catenin pathway in tumor invasion, and provide a conserved mechanism that connects JNK and Wnt signaling in regulating tumor progression.
Collapse
|
228
|
Mo Y, Fan Y, Fu W, Xu W, Chen S, Wen Y, Liu S, Peng L, Xiao Y. Acute immune stress improves cell resistance to chemical poison damage in SP600125-induced polyploidy of fish cells in vitro. FISH & SHELLFISH IMMUNOLOGY 2019; 84:656-663. [PMID: 30393156 DOI: 10.1016/j.fsi.2018.10.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 10/15/2018] [Accepted: 10/24/2018] [Indexed: 06/08/2023]
Abstract
Previous research has indicated that the small compound, SP600125, could induce polyploidy of fish cells, and has established a stable tetraploid cell line from diploid fish cells. In order to explore how fish cells maintain homeostasis under SP600125-stress in vitro, this study investigates impacts of SP600125-stress on intracellular pathways, as well as on regulation of the cellular homeostasis feedback in fish cells. Transcriptomes are obtained from the SP600125-treated cells. Compared with unigenes expressed in control group (crucial carp fin cells), a total of 2670 and 1846 unigenes are significantly upregulated and downregulated in these cells, respectively. Differentially expressed genes are found, which are involved in innate defense, inflammatory pathways and cell adhesion molecules-related pathways. The SP600125-stress enhances cell-mediated immunity, characterized by significantly increasing expression of multiple immune genes. These enhanced immune genes include the pro-inflammatory cytokines (IL-1β, TNF-ɑ, IL-6R), the adaptor signal transducers (STAT, IκBɑ), and the integrins (ɑ2β1, ɑMβ2). Furthermore, mitochondria are contributed to the cellular homeostasis regulation upon the SP600125-stress. The results show that acute inflammation is an adaptive and controlled response to the SP600125-stress, which is beneficial for alleviating toxicity by SP600125. They provide a potential way of breeding fish polyploidy induced by SP600125 in the future research.
Collapse
Affiliation(s)
- Yanxiu Mo
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Basic Medical Science, Xiangnan University, Chenzhou, Hunan, 423000, PR China
| | - Yunpeng Fan
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Wen Fu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Wenting Xu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Shujuan Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Yuanhui Wen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Shaojun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Liangyue Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China.
| | - Yamei Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China.
| |
Collapse
|
229
|
Greenwald EC, Mehta S, Zhang J. Genetically Encoded Fluorescent Biosensors Illuminate the Spatiotemporal Regulation of Signaling Networks. Chem Rev 2018; 118:11707-11794. [PMID: 30550275 PMCID: PMC7462118 DOI: 10.1021/acs.chemrev.8b00333] [Citation(s) in RCA: 316] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cellular signaling networks are the foundation which determines the fate and function of cells as they respond to various cues and stimuli. The discovery of fluorescent proteins over 25 years ago enabled the development of a diverse array of genetically encodable fluorescent biosensors that are capable of measuring the spatiotemporal dynamics of signal transduction pathways in live cells. In an effort to encapsulate the breadth over which fluorescent biosensors have expanded, we endeavored to assemble a comprehensive list of published engineered biosensors, and we discuss many of the molecular designs utilized in their development. Then, we review how the high temporal and spatial resolution afforded by fluorescent biosensors has aided our understanding of the spatiotemporal regulation of signaling networks at the cellular and subcellular level. Finally, we highlight some emerging areas of research in both biosensor design and applications that are on the forefront of biosensor development.
Collapse
Affiliation(s)
- Eric C Greenwald
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Sohum Mehta
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Jin Zhang
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| |
Collapse
|
230
|
Ghose R. Nature of the Pre-Chemistry Ensemble in Mitogen-Activated Protein Kinases. J Mol Biol 2018; 431:145-157. [PMID: 30562484 DOI: 10.1016/j.jmb.2018.12.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/09/2018] [Accepted: 12/10/2018] [Indexed: 10/27/2022]
Abstract
In spite of the availability of a significant amount of structural detail on docking interactions involving mitogen-activated protein kinases (MAPKs) and their substrates, the mechanism by which the disordered phospho-acceptor on the substrate transiently interacts with the kinase catalytic elements and is phosphorylated, often with high efficiency, remains poorly understood. Here, this dynamic interaction is analyzed in the context of available biophysical and biochemical data for ERK2, an archetypal MAPK. A hypothesis about the nature of the ternary complex involving a MAPK, its substrate, and ATP immediately prior to the chemical step (the pre-chemistry complex) is proposed. It is postulated that the solution ensemble (the pre-chemistry ensemble) representing the pre-chemistry complex comprises several conformations that are linked by dynamics on multiple timescales. These individual conformations possess different intrinsic abilities to proceed through the chemical step. The overall rate of chemistry is therefore related to the microscopic nature of the pre-chemistry ensemble, its constituent conformational microstates, and their intrinsic abilities to yield a phosphorylated product. While characterizing these microstates within the pre-chemistry ensemble in atomic or near-atomic detail is an extremely challenging proposition, recent developments in hybrid methodologies that employ computational approaches driven by experimental data appear to provide the most promising path forward toward achieving this goal.
Collapse
Affiliation(s)
- Ranajeet Ghose
- Department of Chemistry and Biochemistry, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA; Graduate Program in Biochemistry, The Graduate Center of CUNY, New York, NY 10016, USA; Graduate Program in Chemistry, The Graduate Center of CUNY, New York, NY 10016, USA; Graduate Program in Physics, The Graduate Center of CUNY, New York, NY 10016, USA
| |
Collapse
|
231
|
Papageorgiou M, Raza A, Fraser S, Nurgali K, Apostolopoulos V. Methamphetamine and its immune-modulating effects. Maturitas 2018; 121:13-21. [PMID: 30704560 DOI: 10.1016/j.maturitas.2018.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022]
Abstract
The recreational use of methamphetamine (METH, or ice) is a global burden. It pervades and plagues contemporary society; it has been estimated that there are up to 35 million users worldwide. METH is a highly addictive psychotropic compound which acts on the central nervous system, and chronic use can induce psychotic behavior. METH has the capacity to modulate immune cells, giving the drug long-term effects which may manifest as neuropsychiatric disorders, and that increase susceptibility to communicable diseases, such as HIV. In addition, changes to the cytokine balance have been associated with compromise of the blood-brain barrier, resulting to alterations to brain plasticity, creating lasting neurotoxicity. Immune-related signaling pathways are key to further evaluating how METH impacts host immunity through these neurological and peripheral modifications. Combining this knowledge with current data on inflammatory responses will improve understanding of how the adaptive and innate immunity responds to METH, how this can activate premature-ageing processes and how METH exacerbates disturbances that lead to non-communicable age-related diseases, including cardiovascular disease, stroke, depression and dementia.
Collapse
Affiliation(s)
- Marco Papageorgiou
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Ali Raza
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Regenerative Medicine and StemCells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia.
| | | |
Collapse
|
232
|
Myette-Côté É, Durrer C, Neudorf H, Bammert TD, Botezelli JD, Johnson JD, DeSouza CA, Little JP. The effect of a short-term low-carbohydrate, high-fat diet with or without postmeal walks on glycemic control and inflammation in type 2 diabetes: a randomized trial. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1210-R1219. [PMID: 30303707 PMCID: PMC6734060 DOI: 10.1152/ajpregu.00240.2018] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022]
Abstract
Lowering carbohydrate consumption effectively lowers glucose, but impacts on inflammation are unclear. The objectives of this study were to: 1) determine whether reducing hyperglycemia by following a low-carbohydrate, high-fat (LC) diet could lower markers of innate immune cell activation in type 2 diabetes (T2D) and 2) examine if the combination of an LC diet with strategically timed postmeal walking was superior to an LC diet alone. Participants with T2D ( n = 11) completed a randomized crossover study involving three 4-day diet interventions: 1) low-fat low-glycemic index (GL), 2) and 3) LC with 15-min postmeal walks (LC+Ex). Four-day mean glucose was significantly lower in the LC+Ex group as compared with LC (-5%, P < 0.05), whereas both LC+Ex (-16%, P < 0.001) and LC (-12%, P < 0.001) conditions were lower than GL. A significant main effect of time was observed for peripheral blood mononuclear cells phosphorylated c-Jun N-terminal kinase ( P < 0.001), with decreases in all three conditions (GL: -32%, LC: -45%, and LC+Ex: -44%). A significant condition by time interaction was observed for monocyte microparticles ( P = 0.040) with a significant decrease in GL (-76%, P = 0.035) and a tendency for a reduction in LC (-70%, P = 0.064), whereas there was no significant change in LC+Ex (0.5%, P = 0.990). Both LC (-27%, P = 0.001) and LC+Ex (-35%, P = 0.005) also led to significant reductions in circulating proinsulin. An LC diet improved 4-day glycemic control and fasting proinsulin levels when compared with GL, with added glucose-lowering benefits when LC was combined with postmeal walking.
Collapse
Affiliation(s)
- Étienne Myette-Côté
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Cody Durrer
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Helena Neudorf
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Tyler D Bammert
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| | - José Diego Botezelli
- Department of Cellular and Physiological Sciences, University of British Columbia , Vancouver, BC , Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, University of British Columbia , Vancouver, BC , Canada
| | - Christopher A DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| |
Collapse
|
233
|
Win S, Than TA, Kaplowitz N. The Regulation of JNK Signaling Pathways in Cell Death through the Interplay with Mitochondrial SAB and Upstream Post-Translational Effects. Int J Mol Sci 2018; 19:ijms19113657. [PMID: 30463289 PMCID: PMC6274687 DOI: 10.3390/ijms19113657] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/17/2018] [Accepted: 11/17/2018] [Indexed: 02/08/2023] Open
Abstract
c-Jun-N-terminal kinase (JNK) activity plays a critical role in modulating cell death, which depends on the level and duration of JNK activation. The kinase cascade from MAPkinase kinase kinase (MAP3K) to MAPkinase kinase (MAP2K) to MAPKinase (MAPK) can be regulated by a number of direct and indirect post-transcriptional modifications, including acetylation, ubiquitination, phosphorylation, and their reversals. Recently, a JNK-mitochondrial SH3-domain binding protein 5 (SH3BP5/SAB)-ROS activation loop has been elucidated, which is required to sustain JNK activity. Importantly, the level of SAB expression in the outer membrane of mitochondria is a major determinant of the set-point for sustained JNK activation. SAB is a docking protein and substrate for JNK, leading to an intramitochondrial signal transduction pathway, which impairs electron transport and promotes reactive oxygen species (ROS) release to sustain the MAPK cascade.
Collapse
Affiliation(s)
- Sanda Win
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Tin Aung Than
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Neil Kaplowitz
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
234
|
Deng L, Gao X, Liu B, He X, Xu J, Qiang J, Wu Q, Liu S. NMT1 inhibition modulates breast cancer progression through stress-triggered JNK pathway. Cell Death Dis 2018; 9:1143. [PMID: 30446635 PMCID: PMC6240078 DOI: 10.1038/s41419-018-1201-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/11/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
Myristoylation is one of key post-translational modifications that involved in signal transduction, cellular transformation and tumorigenesis. Increasing evidence demonstrates that targeting myristoylation might provide a new strategy for eliminating cancers. However, the underlying mechanisms are still yielded unclear. In this study, we demonstrated that genetic inhibition of N-myristoyltransferase NMT1 suppressed initiation, proliferation and invasion of breast cancer cells either in vitro or in vivo. We identified ROS could negatively regulate NMT1 expression and NMT1 knockdown conversely promoted oxidative stress, which formed a feedback loop. Furthermore, inhibition of NMT1 caused degraded proteins increase and ER stress, which cross-talked with mitochondria to produce more ROS. And both of oxidative stress and ER stress could activate JNK pathway, leading to autophagy which abrogated breast cancer progression especially triple-negative breast cancer (TNBC). These studies provide a preclinical proof of concept for targeting NMT1 as a strategy to treat breast cancer.
Collapse
Affiliation(s)
- Lu Deng
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China.,School of Life Science, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xinlei Gao
- School of Life Science, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Bingjie Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China.,School of Life Science, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xueyan He
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China
| | - Jiahui Xu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China
| | - Jiankun Qiang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China
| | - Qingfa Wu
- School of Life Science, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
235
|
Kurtzeborn K, Cebrian C, Kuure S. Regulation of Renal Differentiation by Trophic Factors. Front Physiol 2018; 9:1588. [PMID: 30483151 PMCID: PMC6240607 DOI: 10.3389/fphys.2018.01588] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Classically, trophic factors are considered as proteins which support neurons in their growth, survival, and differentiation. However, most neurotrophic factors also have important functions outside of the nervous system. Especially essential renal growth and differentiation regulators are glial cell line-derived neurotrophic factor (GDNF), bone morphogenetic proteins (BMPs), and fibroblast growth factors (FGFs). Here we discuss how trophic factor-induced signaling contributes to the control of ureteric bud (UB) branching morphogenesis and to maintenance and differentiation of nephrogenic mesenchyme in embryonic kidney. The review includes recent advances in trophic factor functions during the guidance of branching morphogenesis and self-renewal versus differentiation decisions, both of which dictate the control of kidney size and nephron number. Creative utilization of current information may help better recapitulate renal differentiation in vitro, but it is obvious that significantly more basic knowledge is needed for development of regeneration-based renal therapies.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
- GM-Unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
236
|
Mugabo Y, Lim GE. Scaffold Proteins: From Coordinating Signaling Pathways to Metabolic Regulation. Endocrinology 2018; 159:3615-3630. [PMID: 30204866 PMCID: PMC6180900 DOI: 10.1210/en.2018-00705] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/05/2018] [Indexed: 01/13/2023]
Abstract
Among their pleiotropic functions, scaffold proteins are required for the accurate coordination of signaling pathways. It has only been within the past 10 years that their roles in glucose homeostasis and metabolism have emerged. It is well appreciated that changes in the expression or function of signaling effectors, such as receptors or kinases, can influence the development of chronic diseases such as diabetes and obesity. However, little is known regarding whether scaffolds have similar roles in the pathogenesis of metabolic diseases. In general, scaffolds are often underappreciated in the context of metabolism or metabolic diseases. In the present review, we discuss various scaffold proteins and their involvement in signaling pathways related to metabolism and metabolic diseases. The aims of the present review were to highlight the importance of scaffold proteins and to raise awareness of their physiological contributions. A thorough understanding of how scaffolds influence metabolism could aid in the discovery of novel therapeutic approaches to treat chronic conditions, such as diabetes, obesity, and cardiovascular disease, for which the incidence of all continue to increase at alarming rates.
Collapse
Affiliation(s)
- Yves Mugabo
- Cardiometabolic Axis, Centre de Recherche de Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Montréal Diabetes Research Centre, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Gareth E Lim
- Cardiometabolic Axis, Centre de Recherche de Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Montréal Diabetes Research Centre, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
237
|
Rodgers ML, Takeshita R, Griffitt RJ. Deepwater Horizon oil alone and in conjunction with Vibrio anguillarum exposure modulates immune response and growth in red snapper (Lutjanus campechanus). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 204:91-99. [PMID: 30223188 DOI: 10.1016/j.aquatox.2018.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/16/2018] [Accepted: 09/07/2018] [Indexed: 06/08/2023]
Abstract
This study examined the impacts of Macondo oil from the Deepwater Horizon oil spill, both alone and in conjunction with exposure to the known fish pathogen Vibrio anguillarum, on the expression of five immune-related gene transcripts of red snapper (il8, il10, tnfa, il1b, and igm). In order to elucidate this impact, six different test conditions were used: one Control group (No oil/No pathogen), one Low oil/No pathogen group (tPAH50 = 0.563 μg/L), one High oil/No pathogen group (tPAH50 = 17.084 μg/L, one No oil/Pathogen group, one Low oil/Pathogen group (tPAH50 = 0.736 μg/L), and one High oil/Pathogen group (tPAH50 = 15.799 μg/L). Fish were exposed to their respective oil concentrations for one week. On day 7 of the experiment, all fish were placed into new tanks (with or without V. anguillarum) for one hour. At three time points (day 8, day 10, and day 17), fish organs were harvested and placed into RNAlater, and qPCR was run for examination of the above specific immune genes as well as cyp1a1. Our results suggest that cyp1a1 transcripts were upregulated in oil-exposed groups throughout the experiment, confirming oil exposure, and that all five immune gene transcripts were upregulated on day 8, but were generally downregulated or showed no differences from controls on days 10 and 17. Finally, both oil and pathogen exposure had impacts on growth.
Collapse
Affiliation(s)
- Maria L Rodgers
- Division of Coastal Sciences, School of Ocean Science and Technology, University of Southern Mississippi, Ocean Springs, MS, 39564, USA
| | - Ryan Takeshita
- Abt Associates, 1881 Ninth Street, Suite 201, Boulder, CO, USA
| | - Robert J Griffitt
- Division of Coastal Sciences, School of Ocean Science and Technology, University of Southern Mississippi, Ocean Springs, MS, 39564, USA.
| |
Collapse
|
238
|
Ulu A, Frost JA. Regulation of RhoA activation and cell motility by c-Jun N-terminal kinases and Net1. Small GTPases 2018; 11:385-391. [PMID: 30332929 DOI: 10.1080/21541248.2018.1536638] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Jnks are mitogen activated protein kinases that are best known for regulating transcription and apoptotic signaling. However, they also play important roles in controlling cell motility and invasion by phosphorylating many actin and microtubule regulatory proteins. These mechanisms have important implications for normal cell motility as well as cancer metastasis. Jnks are activated by growth factors and cytokines that stimulate cell motility, and this often requires upstream activation of Rho GTPases. Our recent work indicates that Jnks may also regulate Rho GTPase activation. Specifically, we found that Jnk-dependent phosphorylation of the RhoA guanine nucleotide exchange factor (RhoGEF) Net1A promotes its cytosolic accumulation to drive RhoA activation and actin cytoskeletal reorganization. Net1A is unusual among RhoGEFs in that it is sequestered in the nucleus to prevent aberrant RhoA activation. Importantly, Jnk-stimulated cytosolic localization of Net1A is sufficient to stimulate cell motility and extracellular matrix invasion in non-invasive breast cancer cells. Since Net1A expression is critical for cancer cell motility and invasion in vitro, and breast cancer metastasis in vivo, these data uncover a previously unappreciated regulatory mechanism that may contribute to metastasis in multiple types of cancer.
Collapse
Affiliation(s)
- Arzu Ulu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston , Houston, TX, USA
| | - Jeffrey A Frost
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston , Houston, TX, USA
| |
Collapse
|
239
|
Ren F, Mu N, Gao M, Sun J, Zhang C, Sun X, Li L, Li J, Liu T, Tse G, Dong M. Role of JNK signalling pathway and platelet‑lymphocyte aggregates in myocardial ischemia‑reperfusion injury and the cardioprotective effect of ischemic postconditioning in rats. Mol Med Rep 2018; 18:5237-5242. [PMID: 30320401 DOI: 10.3892/mmr.2018.9545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 08/30/2018] [Indexed: 11/06/2022] Open
Abstract
In myocardial ischemia‑reperfusion injury (MIRI), increased activity of the c‑Jun N‑terminal kinase (JNK) pathway and the activation of platelets that leads to the formation of platelet‑leukocyte aggregates (PLAs) have been observed. It was hypothesized that ischemic postconditioning in MIRI exerts cardioprotective effects by altering JNK activity, which in turn leads to reduced PLA levels. A total of 60 rats were randomly divided into 6 groups (n=10 for each group): i) Control; ii) ischemia‑reperfusion injury alone; iii) ischemia‑reperfusion with postconditioning (PostC group), iv) treatment with the JNK inhibitor‑SP600125; v) postC and treatment with anisomycin; and vi) treatment with the JNK activator‑anisomycin. Subsequently, the levels of PLA, infarct size, myocardial injury markers (creatinine kinase‑muscle/brain and troponin I) and were measured. Western blotting was used to determine the protein expression of phosphorylated‑JNK. MIRI led to increased myocardial infarct size that was associated with raised troponin I and creatine kinase‑muscle/brain. At different time points of MIRI, the level of PLA gradually increased. Compared with the injury‑reperfusion group, the level of PLA in the PostC and Inhibitor‑JNK groups was significantly reduced at 60 min and 3 h following reperfusion. MIRI was able to increase the expression of phosphorylated JNK. These effects were significantly reduced by ischemic postC or by treatment with SP600125. By contrast, the addition of anisomycin attenuated these protective effects. JNK is a critical mediator of MIRI. Ischemic postC can reduce the level of PLA during reperfusion by inhibiting the phosphorylation of JNK MAPK, thereby reducing MIRI. Pharmacological inhibition and activation of JNK can improve and reduce cardioprotective effects, respectively. These results explained the mechanism of the cardioprotection of postC and provided novel insight and target for the therapeutic strategy of MIRI.
Collapse
Affiliation(s)
- Faxin Ren
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Nan Mu
- Department of Gynaecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Mingxiao Gao
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Jing Sun
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Chuanhuan Zhang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Xiaojian Sun
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Liudong Li
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Jun Li
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic‑Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Gary Tse
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Mei Dong
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
240
|
Tatsumi Y, Matsumoto N, Iibe N, Watanabe N, Torii T, Sango K, Homma K, Miyamoto Y, Sakagami H, Yamauchi J. CMT type 2N disease-associated AARS mutant inhibits neurite growth that can be reversed by valproic acid. Neurosci Res 2018; 139:69-78. [PMID: 30261202 DOI: 10.1016/j.neures.2018.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/06/2018] [Accepted: 09/17/2018] [Indexed: 12/27/2022]
Abstract
Charcot-Marie-Tooth (CMT) disease is composed of a heterogeneous group of hereditary peripheral neuropathies. The peripheral nervous system primarily comprises two types of cells: neuronal cells and myelinating glial Schwann cells. CMT2 N is an autosomal dominant disease and its responsible gene encodes alanyl-tRNA synthetase (AARS), which is a family of cytoplasmic aminoacyl-tRNA synthetases. CMT2 N is associated with the mutation, including a missense mutation, which is known to decrease the enzymatic activity of AARS, but whether and how its mutation affects AARS localization and neuronal process formation remains to be understood. First, we show that the AARS mutant harboring Asn71-to-Tyr (N71Y) is not localized in cytoplasm. The expression of AARS mutant proteins in COS-7 cells mainly leads to localization into lysosome, whereas the wild type is indeed localized in cytoplasm. Second, in N1E-115 cells as the neuronal cell model, cells expressing the N71Y mutant do not have the ability to grow processes. Third, pretreatment with antiepileptic valproic acid reverses the inhibitory effect of the N71Y mutant on process growth. Taken together, the N71Y mutation of AARS leads to abnormal intracellular localization, inhibiting process growth, yet this inhibition is reversed by valproic acid.
Collapse
Affiliation(s)
- Yuriko Tatsumi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Naoto Matsumoto
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Noriko Iibe
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Natsumi Watanabe
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Tomohiro Torii
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kazunori Sango
- Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, 156-8506, Japan
| | - Keiichi Homma
- Department of Life Science and Informatics, Maebashi Institute of Technology, Maebashi, Gunma, 371-0816, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan; Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo, 157-8535, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0734, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan; Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo, 157-8535, Japan.
| |
Collapse
|
241
|
Walker NM, Mazzoni SM, Vittal R, Fingar DC, Lama VN. c-Jun N-terminal kinase (JNK)-mediated induction of mSin1 expression and mTORC2 activation in mesenchymal cells during fibrosis. J Biol Chem 2018; 293:17229-17239. [PMID: 30217824 DOI: 10.1074/jbc.ra118.003926] [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: 05/15/2018] [Revised: 09/06/2018] [Indexed: 02/03/2023] Open
Abstract
Mammalian target of rapamycin complex 2 (mTORC2) has been shown to regulate mTORC1/4E-BP1/eIF4E signaling and collagen I expression in mesenchymal cells (MCs) during fibrotic activation. Here we investigated the regulation of the mTORC2 binding partner mammalian stress-activated protein kinase-interacting protein 1 (mSin1) in MCs derived from human lung allografts and identified a novel role for mSin1 during fibrosis. mSin1 was identified as a common downstream target of key fibrotic pathways, and its expression was increased in MCs in response to pro-fibrotic mediators: lysophosphatidic acid (LPA), transforming growth factor β, and interleukin 13. Fibrotic MCs had higher mSin1 protein levels than nonfibrotic MCs, and siRNA-mediated silencing of mSIN1 inhibited collagen I expression and mTORC1/2 activity in these cells. Autocrine LPA signaling contributed to constitutive up-regulation of mSin1 in fibrotic MCs, and mSin1 was decreased because of LPA receptor 1 siRNA treatment. We identified c-Jun N-terminal kinase (JNK) as a key intermediary in mSin1 up-regulation by the pro-fibrotic mediators, as pharmacological and siRNA-mediated inhibition of JNK prevented the LPA-induced mSin1 increase. Proteasomal inhibition rescued mSin1 levels after JNK inhibition in LPA-treated MCs, and the decrease in mSin1 ubiquitination in response to LPA was counteracted by JNK inhibitors. Constitutive JNK1 overexpression induced mSin1 expression and could drive mTORC2 and mTORC1 activation and collagen I expression in nonfibrotic MCs, effects that were reversed by siRNA-mediated mSIN1 silencing. These results indicate that LPA stabilizes mSin1 protein expression via JNK signaling by blocking its proteasomal degradation and identify the LPA/JNK/mSin1/mTORC/collagen I pathway as critical for fibrotic activation of MCs.
Collapse
Affiliation(s)
- Natalie M Walker
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and
| | - Serina M Mazzoni
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and
| | - Ragini Vittal
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and
| | - Diane C Fingar
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-0360
| | - Vibha N Lama
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and
| |
Collapse
|
242
|
Nelson KJ, Bolduc JA, Wu H, Collins JA, Burke EA, Reisz JA, Klomsiri C, Wood ST, Yammani RR, Poole LB, Furdui CM, Loeser RF. H 2O 2 oxidation of cysteine residues in c-Jun N-terminal kinase 2 (JNK2) contributes to redox regulation in human articular chondrocytes. J Biol Chem 2018; 293:16376-16389. [PMID: 30190325 DOI: 10.1074/jbc.ra118.004613] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/04/2018] [Indexed: 01/01/2023] Open
Abstract
Reactive oxygen species (ROS), in particular H2O2, regulate intracellular signaling through reversible oxidation of reactive protein thiols present in a number of kinases and phosphatases. H2O2 has been shown to regulate mitogen-activated protein kinase (MAPK) signaling depending on the cellular context. We report here that in human articular chondrocytes, the MAPK family member c-Jun N-terminal kinase 2 (JNK2) is activated by fibronectin fragments and low physiological levels of H2O2 and inhibited by oxidation due to elevated levels of H2O2 The kinase activity of affinity-purified, phosphorylated JNK2 from cultured chondrocytes was reversibly inhibited by 5-20 μm H2O2 Using dimedone-based chemical probes that react specifically with sulfenylated cysteines (RSOH), we identified Cys-222 in JNK2, a residue not conserved in JNK1 or JNK3, as a redox-reactive site. MS analysis of human recombinant JNK2 also detected further oxidation at Cys-222 and other cysteines to sulfinic (RSO2H) or sulfonic (RSO3H) acid. H2O2 treatment of JNK2 resulted in detectable levels of peptides containing intramolecular disulfides between Cys-222 and either Cys-213 or Cys-177, without evidence of dimer formation. Substitution of Cys-222 to alanine rendered JNK2 insensitive to H2O2 inhibition, unlike C177A and C213A variants. Two other JNK2 variants, C116A and C163A, were also resistant to oxidative inhibition. Cumulatively, these findings indicate differential regulation of JNK2 signaling dependent on H2O2 levels and point to key cysteine residues regulating JNK2 activity. As levels of intracellular H2O2 rise, a switch occurs from activation to inhibition of JNK2 activity, linking JNK2 regulation to the redox status of the cell.
Collapse
Affiliation(s)
| | - Jesalyn A Bolduc
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Hanzhi Wu
- the Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and
| | - John A Collins
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Elizabeth A Burke
- the Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and
| | - Julie A Reisz
- the Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and
| | - Chananat Klomsiri
- From the Department of Biochemistry and.,the Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and
| | - Scott T Wood
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Raghunatha R Yammani
- the Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and
| | | | - Cristina M Furdui
- the Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and
| | - Richard F Loeser
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, North Carolina 27599
| |
Collapse
|
243
|
Langfermann DS, Rössler OG, Thiel G. Stimulation of B-Raf increases c-Jun and c-Fos expression and upregulates AP-1-regulated gene transcription in insulinoma cells. Mol Cell Endocrinol 2018; 472:126-139. [PMID: 29225069 DOI: 10.1016/j.mce.2017.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 12/05/2017] [Accepted: 12/07/2017] [Indexed: 01/15/2023]
Abstract
Stimulation of pancreatic β-cells with glucose activates the protein kinases B-Raf and extracellular signal-regulated protein kinase that participate in glucose sensing. Inhibition of both kinases results in impairment of glucose-regulated gene transcription. To analyze the signaling pathway controlled by B-Raf, we expressed a conditionally active form of B-Raf in INS-1 insulinoma cells. Here, we show that stimulation of B-Raf strongly activated the transcription factor AP-1 which is accompanied by increased c-Jun and c-Fos promoter activities, an upregulation of c-Jun and c-Fos biosynthesis, and elevated transcriptional activation potentials of c-Jun and c-Fos. Mutational analysis identified the AP-1 sites within the c-Jun promoter and the serum response element (SRE) within the c-Fos promoter as the essential genetic elements connecting B-Raf stimulation with AP-1 activation. In line with this, the transcriptional activation potential of the SRE-binding protein Elk-1 was increased following B-Raf activation. The signal pathway from B-Raf to AP-1 required the activation of c-Jun. We identified the cyclin D1 gene as a delayed response gene for AP-1 following stimulation of B-Raf in insulinoma cells. Moreover, MAP kinase phosphatase-1 and the Ca2+/calmodulin-dependent protein phosphatase calcineurin were identified to function as shut-off-devices for the signaling cascade connecting B-Raf stimulation with the activation of AP-1. The fact that stimulation with glucose, activation of L-type voltage-gated Ca2+ channels, and stimulation of B-Raf all trigger an activation of AP-1 indicates that AP-1 is a point of convergence of signaling pathways in β-cell.
Collapse
Affiliation(s)
- Daniel S Langfermann
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| | - Oliver G Rössler
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| | - Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany.
| |
Collapse
|
244
|
Dutta D, Singh A, Paul MS, Sharma V, Mutsuddi M, Mukherjee A. Deltex interacts with Eiger and consequently influences the cell death in Drosophila melanogaster. Cell Signal 2018; 49:17-29. [DOI: 10.1016/j.cellsig.2018.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/25/2018] [Accepted: 05/13/2018] [Indexed: 10/16/2022]
|
245
|
Meng LL, Wang JL, Xu SP, Zu LD, Yan ZW, Zhang JB, Han YQ, Fu GH. Low serum gastrin associated with ER + breast cancer development via inactivation of CCKBR/ERK/P65 signaling. BMC Cancer 2018; 18:824. [PMID: 30115027 PMCID: PMC6097285 DOI: 10.1186/s12885-018-4717-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 08/02/2018] [Indexed: 12/24/2022] Open
Abstract
Background Gastrin is an important gastrointestinal hormone produced primarily by G-cells in the antrum of the stomach. It normally regulates gastric acid secretion and is implicated in a number of human disease states, but how its function affects breast cancer (BC) development is not documented. The current study investigated the suppressive effects of gastrin on BC and its underlying mechanisms. Methods Serum levels of gastrin were measured by enzyme-linked immunosorbent assay (ELISA) and correlation between gastrin level and development of BC was analyzed by chi-square test. Inhibitory effects of gastrin on BC were investigated by CCK-8 assay and nude mice models. Expressions of CCKBR/ERK/P65 in BC patients were determined through immunohistochemistry (IHC) and Western blot. Survival analysis was performed using the log-rank test. Results The results indicated that the serum level of gastrin in BC patients was lower compared with normal control. Cellular and molecular experiments indicated that reduction of gastrin is associated with inactivation of cholecystokinin B receptor (CCKBR)/ERK/P65 signaling in BC cells which is corresponding to molecular type of estrogen receptor (ER) positive BC. Furthermore, we found that low expression of gastrin/CCKBR/ERK /P65 was correlated to worse prognosis in BC patients. Gastrin or ERK/P65 activators inhibited ER+ BC through CCKBR-mediated activation of ERK/P65. Moreover, combination treatment with gastrin and tamoxifen more efficiently inhibited ER+ BC than tamoxifen alone. Conclusions We concluded that low serum gastrin is related to increased risk of ER+ BC development. The results also established that CCKBR/ERK/P65 signaling function is generally tumor suppressive in ER+ BC, indicating therapies should focus on restoring, not inhibiting, CCKBR/ERK/P65 pathway activity. Electronic supplementary material The online version of this article (10.1186/s12885-018-4717-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Li Meng
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, People's Republic of China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Long Wang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, People's Republic of China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu-Ping Xu
- Breast Surgery Division, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Li-Dong Zu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, People's Republic of China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhao-Wen Yan
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, People's Republic of China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Bing Zhang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, People's Republic of China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya-Qin Han
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, People's Republic of China
| | - Guo-Hui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, People's Republic of China. .,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
246
|
Abstract
Depression and anxiety are the most common mood disorders affecting 300 million sufferers worldwide. Maladaptive changes in the neuroendocrine stress response is cited as the most common underlying cause, though how the circuits underlying this response are controlled at the molecular level, remains largely unknown. Approximately 40% of patients do not respond to current treatments, indicating that untapped mechanisms exist. Here we review recent evidence implicating JNK in the control of anxiety and depressive-like behavior with a particular focus on its action in immature granule cells of the hippocampal neurogenic niche and the potential for therapeutic targeting for affective disorders.
Collapse
Affiliation(s)
- Patrik Hollos
- Turku Centre for Biotechnology, Åbo Akademi and University of Turku, BioCity, Turku FIN, Finland
| | - Francesca Marchisella
- Turku Centre for Biotechnology, Åbo Akademi and University of Turku, BioCity, Turku FIN, Finland
| | - Eleanor T Coffey
- Turku Centre for Biotechnology, Åbo Akademi and University of Turku, BioCity, Turku FIN, Finland
| |
Collapse
|
247
|
Labriola JM, Zhou Y, Nagar B. Structural Analysis of the Bacterial Effector AvrA Identifies a Critical Helix Involved in Substrate Recognition. Biochemistry 2018; 57:4985-4996. [PMID: 30025209 DOI: 10.1021/acs.biochem.8b00512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bacterial effector proteins are essential for the infection and proliferation of pathogenic bacteria through manipulation of host immune response pathways. AvrA is a Salmonella effector that belongs to the YopJ family of acetyltransferases, which suppresses c-JUN N-terminal kinase (JNK) signaling in mammals through acetylation of mitogen-activated receptor kinase kinases 4 and 7 (MKK4/7). Interestingly, there are two paralogues of AvrA that differ by only a single internal leucine residue, which when absent (AvrAΔL140) abrogates the ability to suppress JNK signaling. Here, we present the first crystal structure of a bacterial effector from an animal pathogen, AvrAΔL140, accompanied by a thorough biophysical characterization of both AvrA variants. The structure in complex with inositol hexaphosphate and coenzyme A reveals two closely associated domains consisting of a catalytic core that resembles the CE clan peptidases and a wedge-shaped regulatory region that mediates cofactor and substrate binding. The loss of the putative function of AvrAΔL140 is due to its inability to interact with MKK4/7, which ultimately arises from an altered conformation of a critical helix adjacent to the active site that harbors L140. These results provide general insights into substrate recognition across the YopJ family of acetyltransferases.
Collapse
Affiliation(s)
- Jonathan M Labriola
- Department of Biochemistry and Groupe de Recherche Axé sur la Structure des Protéines , McGill University , Montreal , QC H3G 0B1 , Canada
| | - Yifan Zhou
- Department of Biochemistry and Groupe de Recherche Axé sur la Structure des Protéines , McGill University , Montreal , QC H3G 0B1 , Canada
| | - Bhushan Nagar
- Department of Biochemistry and Groupe de Recherche Axé sur la Structure des Protéines , McGill University , Montreal , QC H3G 0B1 , Canada
| |
Collapse
|
248
|
Guo J, Cheng Y. RETRACTED: MicroRNA-1247 inhibits lipopolysaccharides-induced acute pneumonia in A549 cells via targeting CC chemokine ligand 16. Biomed Pharmacother 2018; 104:60-68. [DOI: 10.1016/j.biopha.2018.05.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023] Open
|
249
|
Sharma A, Tiwari M, Gupta A, Pandey AN, Yadav PK, Chaube SK. Journey of oocyte from metaphase-I to metaphase-II stage in mammals. J Cell Physiol 2018; 233:5530-5536. [PMID: 29331044 DOI: 10.1002/jcp.26467] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/05/2018] [Indexed: 12/13/2022]
Abstract
In mammals, journey from metaphase-I (M-I) to metaphase-II (M-II) is important since oocyte extrude first polar body (PB-I) and gets converted into haploid gamete. The molecular and cellular changes associated with meiotic cell cycle progression from M-I to M-II stage and extrusion of PB-I remain ill understood. Several factors drive oocyte meiosis from M-I to M-II stage. The mitogen-activated protein kinase3/1 (MAPK3/1), signal molecules and Rho family GTPases act through various pathways to drive cell cycle progression from M-I to M-II stage. The down regulation of MOS/MEK/MAPK3/1 pathway results in the activation of anaphase-promoting complex/cyclosome (APC/C). The active APC/C destabilizes maturation promoting factor (MPF) and induces meiotic resumption. Several signal molecules such as, c-Jun N-terminal kinase (JNK2), SENP3, mitotic kinesin-like protein 2 (MKlp2), regulator of G-protein signaling (RGS2), Epsin2, polo-like kinase 1 (Plk1) are directly or indirectly involved in chromosomal segregation. Rho family GTPase is another enzyme that along with cell division cycle (Cdc42) to form actomyosin contractile ring required for chromosomal segregation. In the presence of origin recognition complex (ORC4), eccentrically localized haploid set of chromosomes trigger cortex differentiation and determine the division site for polar body formation. The actomyosin contractile activity at the site of division plane helps to form cytokinetic furrow that results in the formation and extrusion of PB-I. Indeed, oocyte journey from M-I to M-II stage is coordinated by several factors and pathways that enable oocyte to extrude PB-I. Quality of oocyte directly impact fertilization rate, early embryonic development, and reproductive outcome in mammals.
Collapse
Affiliation(s)
- Alka Sharma
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Meenakshi Tiwari
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Anumegha Gupta
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashutosh N Pandey
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Pramod K Yadav
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shail K Chaube
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
250
|
Kim M, Baek M, Kim DJ. Protein Tyrosine Signaling and its Potential Therapeutic Implications in Carcinogenesis. Curr Pharm Des 2018. [PMID: 28625132 DOI: 10.2174/1381612823666170616082125] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein tyrosine phosphorylation is a crucial signaling mechanism that plays a role in epithelial carcinogenesis. Protein tyrosine kinases (PTKs) control various cellular processes including growth, differentiation, metabolism, and motility by activating major signaling pathways including STAT3, AKT, and MAPK. Genetic mutation of PTKs and/or prolonged activation of PTKs and their downstream pathways can lead to the development of epithelial cancer. Therefore, PTKs became an attractive target for cancer prevention. PTK inhibitors are continuously being developed, and they are currently used for the treatment of cancers that show a high expression of PTKs. Protein tyrosine phosphatases (PTPs), the homeostatic counterpart of PTKs, negatively regulate the rate and duration of phosphotyrosine signaling. PTPs initially were considered to be only housekeeping enzymes with low specificity. However, recent studies have demonstrated that PTPs can function as either tumor suppressors or tumor promoters, depending on their target substrates. Together, both PTK and PTP signal transduction pathways are potential therapeutic targets for cancer prevention and treatment.
Collapse
Affiliation(s)
- Mihwa Kim
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Minwoo Baek
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Dae Joon Kim
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| |
Collapse
|