1
|
Li Z, Gao Z, Sun T, Zhang S, Yang S, Zheng M, Shen H. Meteorin-like/Metrnl, a novel secreted protein implicated in inflammation, immunology, and metabolism: A comprehensive review of preclinical and clinical studies. Front Immunol 2023; 14:1098570. [PMID: 36911663 PMCID: PMC9998503 DOI: 10.3389/fimmu.2023.1098570] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/10/2023] [Indexed: 02/26/2023] Open
Abstract
Meteorin-like, also known as Metrnl, Meteorin-β, Subfatin, and Cometin, is a novel secreted protein exerting pleiotropic effects on inflammation, immunology, and metabolism. Earlier research on this hormone focused on regulating energy expenditure and glucose homeostasis. Consequently, several studies attempted to characterize the molecule mechanism of Metrnl in glucose metabolism and obesity-related disorders but reported contradictory clinical results. Recent studies gradually noticed its multiple protective functions in inflammatory immune regulations and cardiometabolic diseases, such as inducing macrophage activation, angiogenesis, tissue remodeling, bone formation, and preventing dyslipidemias. A comprehensive understanding of this novel protein is essential to identify its significance as a potential therapeutic drug or a biomarker of certain diseases. In this review, we present the current knowledge on the physiology of Metrnl and its roles in inflammation, immunology, and metabolism, including animal/cell interventional preclinical studies and human clinical studies. We also describe controversies regarding the data of circulation Metrnl in different disease states to determine its clinical application better.
Collapse
Affiliation(s)
- Zhuoqi Li
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| | - Ziyu Gao
- Department of Thyroid Surgery, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| | - Tao Sun
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| | - Shipeng Zhang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| | - Shengnan Yang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| | - Meilin Zheng
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| | - Hui Shen
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| |
Collapse
|
2
|
Xu W, Dahlke SP, Sung M, Samal B, Emery AC, Elkahloun A, Eiden LE. ERK-dependent induction of the immediate-early gene Egr1 and the late gene Gpr50 contribute to two distinct phases of PACAP Gs-GPCR signaling for neuritogenesis. J Neuroendocrinol 2022; 34:e13182. [PMID: 35841324 PMCID: PMC9529758 DOI: 10.1111/jne.13182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/28/2022]
Abstract
Gs-coupled GPCR-stimulated neuritogenesis in PC12 and NS-1 - cells depends on activation of the MAP kinase ERK. Here, we examine changes in ERK activation (phosphorylation), and the time course of ERK-dependent gene induction, to seek transcriptional determinants for this process. Quenching of ERK activation by inhibition of MEK with U0126 at any time point for at least 24 h following addition of PACAP resulted in arrest of neurite formation. Changes in the transcriptome profile throughout this time period revealed at least two phases of gene induction: an early phase dominated by induction of immediate-early genes, and a later phase of gene induction after 4-6 h of exposure to PACAP with persistent elevation of phospho-ERK levels. Genes induced by PACAP in both phases consisted in those whose induction was dependent on ERK (i.e., blocked by U0126), and some whose induction was blocked by the protein kinase A inhibitor H89. ERK-dependent "late gene" transcripts included Gpr50, implicated earlier in facilitation of NGF-induced neurite formation in NS-1 cells. Gpr50 induction by PACAP, but not NGF, was dependent on the guanine nucleotide exchange factor RapGEF2, which has been shown to be required for PACAP-induced neuritogenesis in NS-1 cells. Expression of a Gpr50-directed shRNA lowered basal levels of Gpr50 mRNA and attenuated Gpr50 mRNA and GPR50 protein induction by PACAP, with a corresponding attenuation of PACAP-induced neuritogenesis. Gs-GPCR-stimulated neuritogenesis first requires immediate-early gene induction, including that of Egr1 (Zif268/NGF1A/Krox24) as previously reported. This early phase of gene induction, however, is insufficient to maintain the neuritogenic process without ERK-dependent induction of additional late genes, including Gpr50, upon continuous exposure to neurotrophic neuropeptide. Early (Egr1) and late (Gpr50) gene induction by NGF, like that for PACAP, was inhibited by U0126, but was independent of RapGEF2, confirming distinct modes of ERK activation by Gs-coupled GPCRs and neurotrophic tyrosine receptor kinases, converging on a final common ERK-dependent signaling pathway for neuritogenesis.
Collapse
Affiliation(s)
- Wenqin Xu
- Section on Molecular Neuroscience, National Institute of Mental Health-Intramural Research Program
| | - Sam P. Dahlke
- Section on Molecular Neuroscience, National Institute of Mental Health-Intramural Research Program
| | - Michelle Sung
- Section on Molecular Neuroscience, National Institute of Mental Health-Intramural Research Program
| | - Babru Samal
- Section on Molecular Neuroscience, National Institute of Mental Health-Intramural Research Program
| | - Andrew C. Emery
- Section on Molecular Neuroscience, National Institute of Mental Health-Intramural Research Program
| | - Abdel Elkahloun
- Microarray Core, National Human Genome Research Institute, Bethesda, MD, USA
| | - Lee E. Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health-Intramural Research Program
| |
Collapse
|
3
|
Zhang SL, Li ZY, Wang DS, Xu TY, Fan MB, Cheng MH, Miao CY. Aggravated ulcerative colitis caused by intestinal Metrnl deficiency is associated with reduced autophagy in epithelial cells. Acta Pharmacol Sin 2020; 41:763-770. [PMID: 31949292 PMCID: PMC7471395 DOI: 10.1038/s41401-019-0343-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/02/2019] [Indexed: 01/26/2023]
Abstract
Metrnl is a newly identified secreted protein highly expressed in the intestinal epithelium. This study aimed to explore the role and mechanism of intestinal epithelial Metrnl in ulcerative colitis. Metrnl-/- (intestinal epithelial cell-specific Metrnl knockout) mice did not display any phenotypes of colitis under basal conditions. However, under administration of 3% dextran sodium sulfate (DSS) drinking water, colitis was more severe in Metrnl-/- mice than in WT mice, as indicated by comparisons of body weight loss, the presence of occult or gross blood per rectum, stool consistency, shrinkage in the colon, intestinal damage, and serum levels of inflammatory factors. DSS-induced colitis activated autophagy in the colon. This activation was partially inhibited by intestinal epithelial Metrnl deficiency, as indicated by a decrease in Beclin-1 and LC3-II/I and an increase in p62 in DSS-treated Metrnl-/- mice compared with WT mice. These phenomena were further confirmed by observation of autophagosomes and immunofluorescence staining for LC3 in epithelial cells. The autophagy-related AMPK-mTOR-p70S6K pathway was also activated in DSS-induced colitis, and this pathway was partially blocked by intestinal epithelial Metrnl deficiency, as indicated by a decrease in AMPK phosphorylation and an increase in mTOR and p70S6K phosphorylation in DSS-treated Metrnl-/- mice compared with WT mice. Therefore, Metrnl deficiency deteriorated ulcerative colitis at least partially through inhibition of autophagy via the AMPK-mTOR-p70S6K pathway, suggesting that Metrnl is a therapeutic target for ulcerative colitis.
Collapse
|
4
|
Ushach I, Arrevillaga-Boni G, Heller GN, Pone E, Hernandez-Ruiz M, Catalan-Dibene J, Hevezi P, Zlotnik A. Meteorin-like/Meteorin-β Is a Novel Immunoregulatory Cytokine Associated with Inflammation. THE JOURNAL OF IMMUNOLOGY 2018; 201:3669-3676. [PMID: 30464051 DOI: 10.4049/jimmunol.1800435] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022]
Abstract
We have described a novel cytokine encoded by a gene called Meteorin-like (Metrnl). Metrnl is a small (∼28 kDa) secreted protein expressed by activated macrophages and barrier tissues (mucosa and skin). Metrnl production by bone marrow macrophages is induced by several cytokines including TNF-α, IL-17α, IL-12, and IL-4 and inhibited by IFN-γ and TGF-β. Metrnl expression in macrophages is also induced by LPS, and its levels in circulation are associated with inflammatory responses in vivo. Furthermore, Metrnl regulates the production of several cytokines and chemokines in macrophages. We have produced a Metrnl-/- mouse, which is viable and shows normal development. However, it exhibits dysregulated cytokine production, alterations in IgG production, and is highly susceptible to LPS in a sepsis model. Furthermore, older Metrnl-/- mice develop inflammatory lesions, suggesting that Metrnl participates in the control of inflammatory responses. Taken together, these observations indicate that Metrnl encodes a novel immunoregulatory cytokine associated with inflammatory responses that we have designated Meteorin-β.
Collapse
Affiliation(s)
- Irina Ushach
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697.,Institute for Immunology, University of California, Irvine, Irvine, CA 92697; and
| | - Gerardo Arrevillaga-Boni
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697.,Institute for Immunology, University of California, Irvine, Irvine, CA 92697; and
| | - Gina N Heller
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697.,Institute for Immunology, University of California, Irvine, Irvine, CA 92697; and
| | - Egest Pone
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697
| | - Marcela Hernandez-Ruiz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697.,Institute for Immunology, University of California, Irvine, Irvine, CA 92697; and
| | - Jovani Catalan-Dibene
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697.,Institute for Immunology, University of California, Irvine, Irvine, CA 92697; and
| | - Peter Hevezi
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697.,Institute for Immunology, University of California, Irvine, Irvine, CA 92697; and
| | - Albert Zlotnik
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697; .,Institute for Immunology, University of California, Irvine, Irvine, CA 92697; and
| |
Collapse
|
5
|
Involvement of the Urokinase Receptor and Its Endogenous Ligands in the Development of the Brain and the Formation of Cognitive Functions. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s11055-017-0525-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
6
|
Tsuchiya T, Fujii M, Matsuda N, Kunida K, Uda S, Kubota H, Konishi K, Kuroda S. System identification of signaling dependent gene expression with different time-scale data. PLoS Comput Biol 2017; 13:e1005913. [PMID: 29281625 PMCID: PMC5760096 DOI: 10.1371/journal.pcbi.1005913] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/09/2018] [Accepted: 12/01/2017] [Indexed: 01/11/2023] Open
Abstract
Cells decode information of signaling activation at a scale of tens of minutes by downstream gene expression with a scale of hours to days, leading to cell fate decisions such as cell differentiation. However, no system identification method with such different time scales exists. Here we used compressed sensing technology and developed a system identification method using data of different time scales by recovering signals of missing time points. We measured phosphorylation of ERK and CREB, immediate early gene expression products, and mRNAs of decoder genes for neurite elongation in PC12 cell differentiation and performed system identification, revealing the input-output relationships between signaling and gene expression with sensitivity such as graded or switch-like response and with time delay and gain, representing signal transfer efficiency. We predicted and validated the identified system using pharmacological perturbation. Thus, we provide a versatile method for system identification using data with different time scales.
Collapse
Affiliation(s)
- Takaho Tsuchiya
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Masashi Fujii
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Naoki Matsuda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Katsuyuki Kunida
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
- Laboratory of Computational Biology, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Shinsuke Uda
- Division of Integrated Omics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Kubota
- Division of Integrated Omics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Katsumi Konishi
- Department of Computer Science, Faculty of Informatics, Kogakuin University, Tokyo, Japan
| | - Shinya Kuroda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
- CREST, Japan Science and Technology Corporation, Tokyo, Japan
| |
Collapse
|
7
|
Salozhin SV, Borodinova AA, Kvichanskii AA, Mikhailova TV, Spivak YS, Bolshakov AP. An inhibitor of serine proteases, serpinb1a, modulates differentiation of PC12 cells but not cultured neurons. NEUROCHEM J+ 2017. [DOI: 10.1134/s1819712417010123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Ma D, Wako Y. Evaluation of Phenolic Compounds and Neurotrophic/neuroprotective Activity of Cultivar Extracts Derived from Chrysanthemum morifolium Flowers. FOOD SCIENCE AND TECHNOLOGY RESEARCH 2017. [DOI: 10.3136/fstr.23.457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Dongjian Ma
- Graduate School of Mechanical and Biochemical Engineering, Hachinohe Institute of Technology
| | - Yutaka Wako
- Department of Biotechnology and Environmental Engineering, Faculty of Engineering, Hachinohe Institute of Technology
| |
Collapse
|
9
|
Gong W, Liu Y, Wu Z, Wang S, Qiu G, Lin S. Meteorin-Like Shows Unique Expression Pattern in Bone and Its Overexpression Inhibits Osteoblast Differentiation. PLoS One 2016; 11:e0164446. [PMID: 27716826 PMCID: PMC5055347 DOI: 10.1371/journal.pone.0164446] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 09/21/2016] [Indexed: 11/19/2022] Open
Abstract
The present study was performed to identify and characterize genes involved in osteoblasts function. Firstly, we constructed and sequenced a human osteoblast full-length cDNA library to screen for genes whose functions have not been reported and further identify these candidate genes through detecting the relationship with the activator protein-1 (AP-1) transcription factor complex using a dual luciferase reporter system. Only one gene, namely METRNL (Meteorin, glial cell differentiation regulator-like) has been screened out. We performed immunohistochemistry to analyze expression patterns in bone and established a stable transfection MG63 cell line of METRNL-EGFP fusion protein overexpression to analyze the function of METRNL in mineralized nodule formation. Immunohistochemistry showed METRNL expression in hypertrophic chondrocytes and osteoblasts lining trabecular bone surfaces. Overexpression of METRNL inhibited mineralized nodule formation by the MG63 osteosarcoma cell line. Thus, the identified gene, METRNL, which is associated with AP-1 transcription factor complex activity, has a unique expression pattern in bone. In addition, the anomalous expression of METRNL may inhibit bone cell differentiation.
Collapse
Affiliation(s)
- Weiyan Gong
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Yong Liu
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhihong Wu
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shaohai Wang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guixing Qiu
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shouqing Lin
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
10
|
Zheng SL, Li ZY, Song J, Liu JM, Miao CY. Metrnl: a secreted protein with new emerging functions. Acta Pharmacol Sin 2016; 37:571-9. [PMID: 27063217 PMCID: PMC4857552 DOI: 10.1038/aps.2016.9] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/24/2016] [Indexed: 12/15/2022]
Abstract
Secreted proteins play critical roles in physiological and pathological processes and can be used as biomarkers and therapies for aging and disease. Metrnl is a novel secreted protein homologous to the neurotrophin Metrn. But this protein, unlike Metrn that is mainly expressed in the brain, shows a relatively wider distribution in the body with high levels of expression in white adipose tissue and barrier tissues. This protein plays important roles in neural development, white adipose browning and insulin sensitization. Based on its expression and distinct functions, this protein is also called Cometin, Subfatin and Interleukin 39, which refer to its neurotrophic effect, adipokine function and the possible action as a cytokine, respectively. The spectrum of Metrnl functions remains to be determined, and the mechanisms of Metrnl action need to be elucidated. In this review, we focus on the discovery, structural characteristics, expression pattern and physiological functions of Metrnl, which will assist in developing this protein as a new therapeutic target or agent.
Collapse
Affiliation(s)
- Si-li Zheng
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Zhi-yong Li
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Jie Song
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Jian-min Liu
- Stroke Center & Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chao-yu Miao
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100069, China
- E-mail
| |
Collapse
|
11
|
DCLK1 is a broadly dysregulated target against epithelial-mesenchymal transition, focal adhesion, and stemness in clear cell renal carcinoma. Oncotarget 2016; 6:2193-205. [PMID: 25605241 PMCID: PMC4385845 DOI: 10.18632/oncotarget.3059] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/10/2014] [Indexed: 12/29/2022] Open
Abstract
Renal clear cell carcinoma (RCC) is the most common type of kidney cancer and the 8th most common cancer overall in the US. RCC survival rates drop precipitously with regional and distant spread and recent studies have demonstrated that RCC presents an epithelial-mesenchymal transition (EMT) phenotype linked to increased recurrence and decreased survival. EMT is a key characteristic of tumor stem cells (TSCs) along with chemo-resistance and radio-resistance, which are also phenotypic of RCC. Targeting these factors is key to increasing the survival of RCC patients. Doublecortin-like kinase 1 (DCLK1) marks TSCs in pancreatic and colorectal cancer and regulates EMT and stemness. Analysis of the Cancer Genome Atlas' RCC dataset revealed that DCLK1 is overexpressed and dysregulated on the mRNA and epigenetic level in more than 93% of RCC tumors relative to adjacent normal tissue. Immunohistochemistry using α-DCLK1 antibody confirmed overexpression and demonstrated a major increase in immunoreactivity in stage II-III tumors compared to normal kidney and stage I tumors. Small-interfering RNA (siRNA) mediated knockdown of DCLK1 resulted in decreased expression of EMT and pluripotency factors and significantly reduced invasion, migration, focal adhesion, drug-resistance, and clonogenic capacity. These findings suggest that DCLK1 is a novel, overexpressed factor in RCC progression that may be targeted to suppress EMT, metastasis, and stemness in early-stage and advanced RCC to increase patient survival. Moreover, the possibility that DCLK1 may mark a population of tumor stem-like cells in RCC should be further investigated in light of these findings.
Collapse
|
12
|
PACAP enhances axon outgrowth in cultured hippocampal neurons to a comparable extent as BDNF. PLoS One 2015; 10:e0120526. [PMID: 25807538 PMCID: PMC4373823 DOI: 10.1371/journal.pone.0120526] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 01/26/2015] [Indexed: 11/19/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts neurotrophic activities including modulation of synaptic plasticity and memory, hippocampal neurogenesis, and neuroprotection, most of which are shared with brain-derived neurotrophic factor (BDNF). Therefore, the aim of this study was to compare morphological effects of PACAP and BDNF on primary cultured hippocampal neurons. At days in vitro (DIV) 3, PACAP increased neurite length and number to similar levels by BDNF, but vasoactive intestinal polypeptide showed much lower effects. In addition, PACAP increased axon, but not dendrite, length, and soma size at DIV 3 similarly to BDNF. The PACAP antagonist PACAP6–38 completely blocked the PACAP-induced increase in axon, but not dendrite, length. Interestingly, the BDNF-induced increase in axon length was also inhibited by PACAP6–38, suggesting a mechanism involving PACAP signaling. K252a, a TrkB receptor inhibitor, inhibited axon outgrowth induced by PACAP and BDNF without affecting dendrite length. These results indicate that in primary cultured hippocampal neurons, PACAP shows morphological actions via its cognate receptor PAC1, stimulating neurite length and number, and soma size to a comparable extent as BDNF, and that the increase in total neurite length is ascribed to axon outgrowth.
Collapse
|
13
|
Seaborn T, Ravni A, Au R, Chow BKC, Fournier A, Wurtz O, Vaudry H, Eiden LE, Vaudry D. Induction of serpinb1a by PACAP or NGF is required for PC12 cells survival after serum withdrawal. J Neurochem 2014; 131:21-32. [PMID: 24899316 DOI: 10.1111/jnc.12780] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 04/30/2014] [Accepted: 05/13/2014] [Indexed: 01/11/2023]
Abstract
PC12 cells are used to study the signaling mechanisms underlying the neurotrophic and neuroprotective activities of pituitary adenylate cyclase-activating polypeptide (PACAP) and nerve growth factor (NGF). Previous microarray experiments indicated that serpinb1a was the most induced gene after 6 h of treatment with PACAP or NGF. This study confirmed that serpinb1a is strongly activated by PACAP and NGF in a time-dependent manner with a maximum induction (~ 50-fold over control) observed after 6 h of treatment. Co-incubation with PACAP and NGF resulted in a synergistic up-regulation of serpinb1a expression (200-fold over control), suggesting that PACAP and NGF act through complementary mechanisms. Consistently, PACAP-induced serpinb1a expression was not blocked by TrkA receptor inhibition. Nevertheless, the stimulation of serpinb1a expression by PACAP and NGF was significantly reduced in the presence of extracellular signal-regulated kinase, calcineurin, protein kinase A, p38, and PI3K inhibitors, indicating that the two trophic factors share some common pathways in the regulation of serpinb1a. Finally, functional investigations conducted with siRNA revealed that serpinb1a is not involved in the effects of PACAP and NGF on PC12 cell neuritogenesis, proliferation or body cell volume but mediates their ability to block caspases 3/7 activity and to promote PC12 cell survival.
Collapse
Affiliation(s)
- Tommy Seaborn
- Neurotrophic Factor and Neuronal Differentiation Team, Inserm U982, DC2N, Mont-Saint-Aignan, France; International Associated Laboratory Samuel de Champlain, Mont-Saint-Aignan, France; Department of Pediatrics, Hôpital St-François d'Assise, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Laval University, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Weygant N, Qu D, Berry WL, May R, Chandrakesan P, Owen DB, Sureban SM, Ali N, Janknecht R, Houchen CW. Small molecule kinase inhibitor LRRK2-IN-1 demonstrates potent activity against colorectal and pancreatic cancer through inhibition of doublecortin-like kinase 1. Mol Cancer 2014; 13:103. [PMID: 24885928 PMCID: PMC4030036 DOI: 10.1186/1476-4598-13-103] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 04/24/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Doublecortin-like kinase 1 (DCLK1) is emerging as a tumor specific stem cell marker in colorectal and pancreatic cancer. Previous in vitro and in vivo studies have demonstrated the therapeutic effects of inhibiting DCLK1 with small interfering RNA (siRNA) as well as genetically targeting the DCLK1+ cell for deletion. However, the effects of inhibiting DCLK1 kinase activity have not been studied directly. Therefore, we assessed the effects of inhibiting DCLK1 kinase activity using the novel small molecule kinase inhibitor, LRRK2-IN-1, which demonstrates significant affinity for DCLK1. RESULTS Here we report that LRRK2-IN-1 demonstrates potent anti-cancer activity including inhibition of cancer cell proliferation, migration, and invasion as well as induction of apoptosis and cell cycle arrest. Additionally we found that it regulates stemness, epithelial-mesenchymal transition, and oncogenic targets on the molecular level. Moreover, we show that LRRK2-IN-1 suppresses DCLK1 kinase activity and downstream DCLK1 effector c-MYC, and demonstrate that DCLK1 kinase activity is a significant factor in resistance to LRRK2-IN-1. CONCLUSIONS Given DCLK1's tumor stem cell marker status, a strong understanding of its biological role and interactions in gastrointestinal tumors may lead to discoveries that improve patient outcomes. The results of this study suggest that small molecule inhibitors of DCLK1 kinase should be further investigated as they may hold promise as anti-tumor stem cell drugs.
Collapse
Affiliation(s)
- Nathaniel Weygant
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Dongfeng Qu
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - William L Berry
- Peggy and Charles Stephenson Oklahoma Cancer Center, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Randal May
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | | | - Daniel B Owen
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sripathi M Sureban
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Naushad Ali
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Peggy and Charles Stephenson Oklahoma Cancer Center, Oklahoma City, OK, USA
| | - Ralf Janknecht
- Peggy and Charles Stephenson Oklahoma Cancer Center, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Courtney W Houchen
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Peggy and Charles Stephenson Oklahoma Cancer Center, Oklahoma City, OK, USA
- COARE Biotechnology, Oklahoma City, OK, USA
| |
Collapse
|
15
|
Zhang K, Duan L, Ong Q, Lin Z, Varman PM, Sung K, Cui B. Light-mediated kinetic control reveals the temporal effect of the Raf/MEK/ERK pathway in PC12 cell neurite outgrowth. PLoS One 2014; 9:e92917. [PMID: 24667437 PMCID: PMC3965503 DOI: 10.1371/journal.pone.0092917] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 02/26/2014] [Indexed: 11/29/2022] Open
Abstract
It has been proposed that differential activation kinetics allows cells to use a common set of signaling pathways to specify distinct cellular outcomes. For example, nerve growth factor (NGF) and epidermal growth factor (EGF) induce different activation kinetics of the Raf/MEK/ERK signaling pathway and result in differentiation and proliferation, respectively. However, a direct and quantitative linkage between the temporal profile of Raf/MEK/ERK activation and the cellular outputs has not been established due to a lack of means to precisely perturb its signaling kinetics. Here, we construct a light-gated protein-protein interaction system to regulate the activation pattern of the Raf/MEK/ERK signaling pathway. Light-induced activation of the Raf/MEK/ERK cascade leads to significant neurite outgrowth in rat PC12 pheochromocytoma cell lines in the absence of growth factors. Compared with NGF stimulation, light stimulation induces longer but fewer neurites. Intermittent on/off illumination reveals that cells achieve maximum neurite outgrowth if the off-time duration per cycle is shorter than 45 min. Overall, light-mediated kinetic control enables precise dissection of the temporal dimension within the intracellular signal transduction network.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Liting Duan
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Qunxiang Ong
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Ziliang Lin
- Department of Applied Physics, Stanford University, Stanford, California, United States of America
| | - Pooja Mahendra Varman
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Kijung Sung
- Biophysics Program, Stanford University, Stanford, California, United States of America
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
16
|
Li ZY, Zheng SL, Wang P, Xu TY, Guan YF, Zhang YJ, Miao CY. Subfatin is a novel adipokine and unlike Meteorin in adipose and brain expression. CNS Neurosci Ther 2014; 20:344-54. [PMID: 24393292 DOI: 10.1111/cns.12219] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 11/25/2013] [Accepted: 11/29/2013] [Indexed: 12/26/2022] Open
Abstract
AIMS Adipose tissue releases adipokines that play important roles in metabolic and cardio-cerebro-vascular homeostasis. This study was to discover novel adipokines using caloric restriction model. METHODS Adipokine candidates were captured by gene array and bioinformatics analysis and verified by preparation of recombinant protein and antibody. RESULTS We established a potential secreted protein database containing 208 genes and identified a novel adipokine, Subfatin, that was the highest expressed in subcutaneous fat of both rodents and humans among 15 detected tissues. The secreted mammalian Subfatin was a glycosylated protein. Subfatin was located diffusely throughout the adipose tissue except lipid droplets, with comparable expression between adipocytes and stromal cells, but much lower expression in macrophages than adipocytes. Subfatin was downregulated in white adipose tissue of caloric restriction rats, whereas dramatically upregulated during white adipocyte differentiation as well as in white adipose tissue of diet-induced obese mice. Subfatin was annotated as Meteorin-like (Metrnl) in public databases, a similar transcript of Meteorin (Metrn, also known as glial cell differentiation regulator). Meteorin displayed a brain-specific expression and was scarce in various adipose tissues, in contrast to the tissue expression patterns of Subfatin. CONCLUSIONS Subfatin is a novel adipokine regulated by adipogenesis and obesity, with tissue distribution different from its homologue Meteorin.
Collapse
Affiliation(s)
- Zhi-Yong Li
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Akimoto Y, Yugi K, Uda S, Kudo T, Komori Y, Kubota H, Kuroda S. The extraction of simple relationships in growth factor-specific multiple-input and multiple-output systems in cell-fate decisions by backward elimination PLS regression. PLoS One 2013; 8:e72780. [PMID: 24039801 PMCID: PMC3767677 DOI: 10.1371/journal.pone.0072780] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/12/2013] [Indexed: 12/31/2022] Open
Abstract
Cells use common signaling molecules for the selective control of downstream gene expression and cell-fate decisions. The relationship between signaling molecules and downstream gene expression and cellular phenotypes is a multiple-input and multiple-output (MIMO) system and is difficult to understand due to its complexity. For example, it has been reported that, in PC12 cells, different types of growth factors activate MAP kinases (MAPKs) including ERK, JNK, and p38, and CREB, for selective protein expression of immediate early genes (IEGs) such as c-FOS, c-JUN, EGR1, JUNB, and FOSB, leading to cell differentiation, proliferation and cell death; however, how multiple-inputs such as MAPKs and CREB regulate multiple-outputs such as expression of the IEGs and cellular phenotypes remains unclear. To address this issue, we employed a statistical method called partial least squares (PLS) regression, which involves a reduction of the dimensionality of the inputs and outputs into latent variables and a linear regression between these latent variables. We measured 1,200 data points for MAPKs and CREB as the inputs and 1,900 data points for IEGs and cellular phenotypes as the outputs, and we constructed the PLS model from these data. The PLS model highlighted the complexity of the MIMO system and growth factor-specific input-output relationships of cell-fate decisions in PC12 cells. Furthermore, to reduce the complexity, we applied a backward elimination method to the PLS regression, in which 60 input variables were reduced to 5 variables, including the phosphorylation of ERK at 10 min, CREB at 5 min and 60 min, AKT at 5 min and JNK at 30 min. The simple PLS model with only 5 input variables demonstrated a predictive ability comparable to that of the full PLS model. The 5 input variables effectively extracted the growth factor-specific simple relationships within the MIMO system in cell-fate decisions in PC12 cells.
Collapse
Affiliation(s)
- Yuki Akimoto
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Katsuyuki Yugi
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Shinsuke Uda
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Takamasa Kudo
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Yasunori Komori
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Hiroyuki Kubota
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Shinya Kuroda
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- Department of Computational Biology, Graduate School of Frontier Sciences, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- CREST, Japan Science and Technology Corporation, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
18
|
de Cubas AA, Leandro-García LJ, Schiavi F, Mancikova V, Comino-Méndez I, Inglada-Pérez L, Perez-Martinez M, Ibarz N, Ximénez-Embún P, López-Jiménez E, Maliszewska A, Letón R, Gómez Graña A, Bernal C, Alvarez-Escolá C, Rodríguez-Antona C, Opocher G, Muñoz J, Megias D, Cascón A, Robledo M. Integrative analysis of miRNA and mRNA expression profiles in pheochromocytoma and paraganglioma identifies genotype-specific markers and potentially regulated pathways. Endocr Relat Cancer 2013; 20:477-93. [PMID: 23660872 DOI: 10.1530/erc-12-0183] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pheochromocytomas (PCCs) and paragangliomas (PGLs) are rare neuroendocrine neoplasias of neural crest origin that can be part of several inherited syndromes. Although their mRNA profiles are known to depend on genetic background, a number of questions related to tumor biology and clinical behavior remain unanswered. As microRNAs (miRNAs) are key players in the modulation of gene expression, their comprehensive analysis could resolve some of these issues. Through characterization of miRNA profiles in 69 frozen tumors with germline mutations in the genes SDHD, SDHB, VHL, RET, NF1, TMEM127, and MAX, we identified miRNA signatures specific to, as well as common among, the genetic groups of PCCs/PGLs. miRNA expression profiles were validated in an independent series of 30 composed of VHL-, SDHB-, SDHD-, and RET-related formalin-fixed paraffin-embedded PCC/PGL samples using quantitative real-time PCR. Upregulation of miR-210 in VHL- and SDHB-related PCCs/PGLs was verified, while miR-137 and miR-382 were confirmed as generally upregulated in PCCs/PGLs (except in MAX-related tumors). Also, we confirmed overexpression of miR-133b as VHL-specific miRNAs, miR-488 and miR-885-5p as RET-specific miRNAs, and miR-183 and miR-96 as SDHB-specific miRNAs. To determine the potential roles miRNAs play in PCC/PGL pathogenesis, we performed bioinformatic integration and pathway analysis using matched mRNA profiling data that indicated a common enrichment of pathways associated with neuronal and neuroendocrine-like differentiation. We demonstrated that miR-183 and/or miR-96 impede NGF-induced differentiation in PC12 cells. Finally, global proteomic analysis in SDHB and MAX tumors allowed us to determine that miRNA regulation occurs primarily through mRNA degradation in PCCs/PGLs, which partially confirmed our miRNA-mRNA integration results.
Collapse
Affiliation(s)
- Aguirre A de Cubas
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Centro Nacional de Investigaciones Oncológicas, Melchor Fernández Almagro 3, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Vezyraki A, Kapelouzouc S, Fotiadisb N, Theofilogiannakosa MS, Gerou E. WITHDRAWN: Identification of meteorin and metrnl as two novel pro-differentiative adipokines: Possible roles in controlling adipogenesis and insulin sensitivity. Biochem Biophys Res Commun 2013:S0006-291X(13)01148-0. [PMID: 23860268 DOI: 10.1016/j.bbrc.2013.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 07/04/2013] [Indexed: 06/02/2023]
Abstract
BBRC has been targeted by a scheme to defraud our editors, reviewers and readers with submission of a manuscript with falsified author and institutional information and therefore wholly unverifiable scientific claims. The manuscript has been withdrawn. We consider such abuse of the editorial and peer review system with the submission of fictional content unethical and it wastes the valuable time of all those who contributed to the evaluation of this manuscript. We are currently exploring which local authorities would have jurisdiction, and will with such authorities explore the question of whether this also constitutes a criminal case of internet fraud and we anticipate turning over to them all of the information we have been able to attain from EES regarding the source of the fraudulent submission.
Collapse
Affiliation(s)
- Alkistis Vezyraki
- School of Health Sciences, University of Thessaly, Karies, Trikala, Greece
| | | | | | | | | |
Collapse
|
20
|
Saito TH, Uda S, Tsuchiya T, Ozaki YI, Kuroda S. Temporal decoding of MAP kinase and CREB phosphorylation by selective immediate early gene expression. PLoS One 2013; 8:e57037. [PMID: 23469182 PMCID: PMC3587639 DOI: 10.1371/journal.pone.0057037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/16/2013] [Indexed: 12/23/2022] Open
Abstract
A wide range of growth factors encode information into specific temporal patterns of MAP kinase (MAPK) and CREB phosphorylation, which are further decoded by expression of immediate early gene products (IEGs) to exert biological functions. However, the IEG decoding system remain unknown. We built a data-driven based on time courses of MAPK and CREB phosphorylation and IEG expression in response to various growth factors to identify how signal is processed. We found that IEG expression uses common decoding systems regardless of growth factors and expression of each IEG differs in upstream dependency, switch-like response, and linear temporal filters. Pulsatile ERK phosphorylation was selectively decoded by expression of EGR1 rather than c-FOS. Conjunctive NGF and PACAP stimulation was selectively decoded by synergistic JUNB expression through switch-like response to c-FOS. Thus, specific temporal patterns and combinations of MAPKs and CREB phosphorylation can be decoded by selective IEG expression via distinct temporal filters and switch-like responses. The data-driven modeling is versatile for analysis of signal processing and does not require detailed prior knowledge of pathways.
Collapse
Affiliation(s)
- Takeshi H. Saito
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shinsuke Uda
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takaho Tsuchiya
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yu-ichi Ozaki
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shinya Kuroda
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
- CREST, Japan Science and Technology Corporation, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|