1
|
Lechuga S, Marino-Melendez A, Davis A, Zalavadia A, Khan A, Longworth MS, Ivanov AI. Coactosin-like protein 1 regulates integrity and repair of model intestinal epithelial barriers via actin binding dependent and independent mechanisms. Front Cell Dev Biol 2024; 12:1405454. [PMID: 39040043 PMCID: PMC11260685 DOI: 10.3389/fcell.2024.1405454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
The actin cytoskeleton regulates the integrity and repair of epithelial barriers by mediating the assembly of tight junctions (TJs), and adherens junctions (AJs), and driving epithelial wound healing. Actin filaments undergo a constant turnover guided by numerous actin-binding proteins, however, the roles of actin filament dynamics in regulating intestinal epithelial barrier integrity and repair remain poorly understood. Coactosin-like protein 1 (COTL1) is a member of the ADF/cofilin homology domain protein superfamily that binds and stabilizes actin filaments. COTL1 is essential for neuronal and cancer cell migration, however, its functions in epithelia remain unknown. The goal of this study is to investigate the roles of COTL1 in regulating the structure, permeability, and repair of the epithelial barrier in human intestinal epithelial cells (IEC). COTL1 was found to be enriched at apical junctions in polarized IEC monolayers in vitro. The knockdown of COTL1 in IEC significantly increased paracellular permeability, impaired the steady state TJ and AJ integrity, and attenuated junctional reassembly in a calcium-switch model. Consistently, downregulation of COTL1 expression in Drosophila melanogaster increased gut permeability. Loss of COTL1 attenuated collective IEC migration and decreased cell-matrix attachment. The observed junctional abnormalities in COTL1-depleted IEC were accompanied by the impaired assembly of the cortical actomyosin cytoskeleton. Overexpression of either wild-type COTL1 or its actin-binding deficient mutant tightened the paracellular barrier and activated junction-associated myosin II. Furthermore, the actin-uncoupled COTL1 mutant inhibited epithelial migration and matrix attachment. These findings highlight COTL1 as a novel regulator of the intestinal epithelial barrier integrity and repair.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
2
|
Wang X, Bai Y, Wang B. Coactosin-Like Protein 1 (COTL1) Could Be an Immunological and Prognostic Biomarker: From Pan-Cancer Analysis to Low-Grade Glioma Validation. J Inflamm Res 2024; 17:1805-1820. [PMID: 38523681 PMCID: PMC10960547 DOI: 10.2147/jir.s453509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
Background Cancer represents a widespread global health challenge impacting millions of individuals worldwide. Identifying new targets for cancer treatment is a crucial step in developing more effective therapies. Among these potential targets, Coactosin-like protein 1 (COTL1), a cytoskeleton-associated protein with critical roles in cell migration, adhesion, and signaling, has shown involvement in tumor progression. Methods GSCA, TIMER, SangerBox database were used to explore the COTL1 expression across different tumor types. We employed the TCGA Pan-Atlas Cancer Genomics Dataset, which is available through the cBioportal platform, to explore genetic alterations in COTL1. We conduct a comprehensive analysis of COTL1, encompassing gene expression, clinical prognosis, RNA modification, immunotherapy, and cancer stemness through SangerBox database. Clinical samples were validated using immunohistochemistry. Results Our analysis revealed that COTL1 is highly expressed in most cancers and correlates with decreased survival in Glioma, Glioblastoma multiforme, and pan-kidney cohorts. Furthermore, COTL1 was found to be associated with DNA and RNA stemness in 20 and 22 different tumor types, respectively. Additionally, COTL1 showed positive correlations with immunological checkpoints and immune infiltration cells. It was also linked to tumor mutation burden (TMB), microsatellite instability (MSI), neoantigen (NEO), and programmed death ligand 1 (PD-L1), all of which are potential targets for immunotherapies. Moreover, a favorable relationship was demonstrated between genomic-instability markers such as heterozygosity (LOH), homologous recombination deficiency (HRD), and mutant allele tumor heterogeneity (MATH) with COTL1. Furthermore, our findings confirmed a positive correlation between COTL1 expression, CD8, and PD-L1 in LGG, as well as an association of high COTL1 expression with decreased patient survival in LGG. Conclusion Based on these compelling findings, COTL1 may hold significant clinical implications for the development of novel cancer therapies and serve as a potential target for tumors associated with immunotherapy in the future.
Collapse
Affiliation(s)
- Xiaoyun Wang
- Institute of Integration of Traditional Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, People’s Republic of China
- Wuxi School of Medicine, Jiangnan University, Wuxi, People’s Republic of China
| | - Yuwei Bai
- Institute of Integration of Traditional Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, People’s Republic of China
- Wuxi School of Medicine, Jiangnan University, Wuxi, People’s Republic of China
| | - Bei Wang
- Institute of Integration of Traditional Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, People’s Republic of China
- Wuxi School of Medicine, Jiangnan University, Wuxi, People’s Republic of China
| |
Collapse
|
3
|
Glathar AR, Oyelakin A, Nayak KB, Sosa J, Romano RA, Sinha S. A Systemic and Integrated Analysis of p63-Driven Regulatory Networks in Mouse Oral Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:446. [PMID: 36672394 PMCID: PMC9856320 DOI: 10.3390/cancers15020446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/04/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignancy of the oral cavity and is linked to tobacco exposure, alcohol consumption, and human papillomavirus infection. Despite therapeutic advances, a lack of molecular understanding of disease etiology, and delayed diagnoses continue to negatively affect survival. The identification of oncogenic drivers and prognostic biomarkers by leveraging bulk and single-cell RNA-sequencing datasets of OSCC can lead to more targeted therapies and improved patient outcomes. However, the generation, analysis, and continued utilization of additional genetic and genomic tools are warranted. Tobacco-induced OSCC can be modeled in mice via 4-nitroquinoline 1-oxide (4NQO), which generates a spectrum of neoplastic lesions mimicking human OSCC and upregulates the oncogenic master transcription factor p63. Here, we molecularly characterized established mouse 4NQO treatment-derived OSCC cell lines and utilized RNA and chromatin immunoprecipitation-sequencing to uncover the global p63 gene regulatory and signaling network. We integrated our p63 datasets with published bulk and single-cell RNA-sequencing of mouse 4NQO-treated tongue and esophageal tumors, respectively, to generate a p63-driven gene signature that sheds new light on the role of p63 in murine OSCC. Our analyses reveal known and novel players, such as COTL1, that are regulated by p63 and influence various oncogenic processes, including metastasis. The identification of new sets of potential biomarkers and pathways, some of which are functionally conserved in human OSCC and can prognosticate patient survival, offers new avenues for future mechanistic studies.
Collapse
Affiliation(s)
- Alexandra Ruth Glathar
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Akinsola Oyelakin
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Kasturi Bala Nayak
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Jennifer Sosa
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
4
|
Tonner H, Hunn S, Auler N, Schmelter C, Pfeiffer N, Grus FH. Dynamin-like Protein 1 (DNML1) as a Molecular Target for Antibody-Based Immunotherapy to Treat Glaucoma. Int J Mol Sci 2022; 23:ijms232113618. [PMID: 36362420 PMCID: PMC9654827 DOI: 10.3390/ijms232113618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/23/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022] Open
Abstract
Slow and progressive loss of retinal ganglion cells (RGCs) is the main characteristic of glaucoma, the second leading cause of blindness worldwide. Previous studies have shown that impaired mitochondrial dynamics could facilitate retinal neurodegeneration. Mitochondrial dynamics are regulated directly (fission) or more indirectly (fusion) by dynamin-like protein 1 (DNML1). Therefore, DNM1L might be a promising target for an antibody-based approach to treat glaucoma. The consequences of targeting endogenous DNM1L by antibodies in a glaucoma animal model have not been investigated yet. Here, we show that the intravitreal application of an anti-DNM1L antibody showed protective effects regarding the survival of RGCs and their axons in the retinal nerve fiber layer (RNFL). Antibody treatment also improved retinal functionality, as observed by electroretinography (Ganzfeld ERG). Western blot analysis revealed altered DNM1L phosphorylation and altered expression of proteins related to apoptosis suggesting a decreased apoptosis rate. Mass spectrometry analysis revealed 28 up-regulated and 21 down-regulated proteins (p < 0.05) in both experimental groups. Protein pathway analysis showed that many proteins interacted directly with the target protein DNM1L and could be classified into three main protein clusters: Vesicle traffic-associated (NSF, SNCA, ARF1), mitochondrion-associated (HSP9A, SLC25A5/ANT2, GLUD1) and cytoskeleton-associated (MAP1A) signaling pathway. Our results demonstrate that DNM1L is a promising target for an antibody-based approach to glaucoma therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Franz H. Grus
- Correspondence: ; Tel.: +49-6131-17-3328; Fax: +49-6131-4970563
| |
Collapse
|
5
|
Zhang F, Fu X, Kataoka M, Liu N, Wang Y, Gao F, Liang T, Dong X, Pei J, Hu X, Zhu W, Yu H, Cowan DB, Hu X, Huang ZP, Wang J, Wang DZ, Chen J. Long noncoding RNA Cfast regulates cardiac fibrosis. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 23:377-392. [PMID: 33473324 PMCID: PMC7787992 DOI: 10.1016/j.omtn.2020.11.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
Cardiac fibrosis occurs in most cardiac diseases, which reduces cardiac muscle compliance, impairs both systolic and diastolic heart function and, ultimately, leads to heart failure. Long noncoding RNAs (lncRNAs) have recently emerged as important regulators of a variety of biological processes; however, little is known about the expression and function of lncRNAs in cardiac fibrosis. Using unbiased transcriptome profiling in a mouse model of myocardial infarction (MI), we identified a cardiac fibroblast-enriched lncRNA (AK048087) named cardiac fibroblast-associated transcript (Cfast), which is significantly elevated after MI. Silencing Cfast expression by small interfering RNAs (siRNAs) or lentiviral short hairpin RNAs (shRNAs) resulted in suppression of fibrosis-related gene expression and transdifferentiation of myofibroblasts into cardiac fibroblasts. Depletion of Cfast by lentiviral shRNAs in mouse hearts significantly attenuated cardiac fibrosis induced by MI or isoproterenol-infusion. Importantly, inhibition of Cfast ameliorated cardiac function following cardiac injury. RNA pull-down followed by mass spectrometry analyses identified COTL1 (coactosin-like 1) as one of the Cfast interacting proteins. Mechanistically, Cfast competitively inhibits the COTL1 interaction with TRAP1 (transforming growth factor-β receptor-associated protein 1), which enhances TGF-β signaling by augmenting SMAD2/SMAD4 complex formation. Therefore, our study identifies Cfast as a novel cardiac fibroblast-enriched lncRNA that regulates cardiac fibroblast activation in response to pathophysiological stress. Cfast could serve as a potential therapeutic target for the prevention of cardiac fibrosis and cardiac diseases.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Xuyang Fu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Masaharu Kataoka
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Ning Liu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Yingchao Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Gao
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Tian Liang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Xiaoxuan Dong
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Jianqiu Pei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Wei Zhu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Hong Yu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Douglas B Cowan
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Xinyang Hu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian'an Wang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jinghai Chen
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| |
Collapse
|
6
|
Shao S, Fan Y, Zhong C, Zhu X, Zhu J. Coactosin-Like Protein (COTL1) Promotes Glioblastoma (GBM) Growth in vitro and in vivo. Cancer Manag Res 2020; 12:10909-10917. [PMID: 33154670 PMCID: PMC7608606 DOI: 10.2147/cmar.s246030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 09/23/2020] [Indexed: 11/23/2022] Open
Abstract
Objective To assess the expression levels of COTL1 in human GBM tissues and evaluate the potential involvement of COTL1 in cancer progression. Methods Bioinformation analysis was performed to evaluate COTL1 mRNA levels in GBM tissues and normal tissues, according to the TCGA database, and explore the effects on prognosis. Immunohistochemical (IHC) assays were performed to evaluate COTL1 expression in human GBM tissues and the clinical pathological analysis was performed. Colony formation and MTT assays were performed to evaluate the effects of COTL1 on GBM cell proliferation. Immunoblot assays were performed to detect the expression level of COTL1, Ki67, and PCNA. A xenograft model was developed in mice to assess the effects of COTL1 on tumor growth in vivo. Results We found COTL1 had an obvious high expression in human GBM tissues. The expression of COTL1 was related to recurrence (P=0.006**) and prognosis of patients with GBM. Our data further demonstrated COTL1 promoted cell proliferation in vitro and contributed to tumor growth of GBM cells in mice. Conclusion We therefore identified a novel and promising therapeutic target for the treatment of GBM.
Collapse
Affiliation(s)
- Shike Shao
- Department of Neurosurgery, The Second People's Hospital of Lianyungang City, Jiangsu, People's Republic of China
| | - Yongjun Fan
- Department of Neurosurgery, The Second People's Hospital of Lianyungang City, Jiangsu, People's Republic of China
| | - Chongpei Zhong
- Department of Neurosurgery, The Second People's Hospital of Lianyungang City, Jiangsu, People's Republic of China
| | - Xianlong Zhu
- Department of Neurosurgery, The Second People's Hospital of Lianyungang City, Jiangsu, People's Republic of China
| | - Jiaqiu Zhu
- Department of Neurosurgery, The Second People's Hospital of Lianyungang City, Jiangsu, People's Republic of China
| |
Collapse
|
7
|
Tan DX, Chen XX, Bai TZ, Zhang J, Li ZF. RETRACTED: Sevoflurane up-regulates microRNA-204 to ameliorate myocardial ischemia/reperfusion injury in mice by suppressing Cotl1. Life Sci 2020; 259:118162. [PMID: 32730836 DOI: 10.1016/j.lfs.2020.118162] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figures 4C+E and 7E, which appear to have a similar phenotype as seen in many other publications, as detailed here: https://pubpeer.com/publications/CE1E814DD630D160BEEBFC2842FE45; and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. The journal requested that the corresponding author comment on these concerns and provide the raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Dian-Xiang Tan
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| | - Xiao-Xi Chen
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| | - Tai-Zhu Bai
- Department of Cardiovascular Medicine, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| | - Juan Zhang
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| | - Zhen-Fa Li
- Department of General Surgery, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| |
Collapse
|
8
|
Kazemi T, Huang S, Avci NG, Waits CMK, Akay YM, Akay M. Investigating the influence of perinatal nicotine and alcohol exposure on the genetic profiles of dopaminergic neurons in the VTA using miRNA-mRNA analysis. Sci Rep 2020; 10:15016. [PMID: 32929144 PMCID: PMC7490691 DOI: 10.1038/s41598-020-71875-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
Nicotine and alcohol are two of the most commonly used and abused recreational drugs, are often used simultaneously, and have been linked to significant health hazards. Furthermore, patients diagnosed with dependence on one drug are highly likely to be dependent on the other. Several studies have shown the effects of each drug independently on gene expression within many brain regions, including the ventral tegmental area (VTA). Dopaminergic (DA) neurons of the dopamine reward pathway originate from the VTA, which is believed to be central to the mechanism of addiction and drug reinforcement. Using a well-established rat model for both nicotine and alcohol perinatal exposure, we investigated miRNA and mRNA expression of dopaminergic (DA) neurons of the VTA in rat pups following perinatal alcohol and joint nicotine-alcohol exposure. Microarray analysis was then used to profile the differential expression of both miRNAs and mRNAs from DA neurons of each treatment group to further explore the altered genes and related biological pathways modulated. Predicted and validated miRNA-gene target pairs were analyzed to further understand the roles of miRNAs within these networks following each treatment, along with their post transcription regulation points affecting gene expression throughout development. This study suggested that glutamatergic synapse and axon guidance pathways were specifically enriched and many miRNAs and genes were significantly altered following alcohol or nicotine-alcohol perinatal exposure when compared to saline control. These results provide more detailed insight into the cell proliferation, neuronal migration, neuronal axon guidance during the infancy in rats in response to perinatal alcohol/ or nicotine-alcohol exposure.
Collapse
Affiliation(s)
- Tina Kazemi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Shuyan Huang
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Naze G Avci
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Charlotte Mae K Waits
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
9
|
Hasan MM, Teixeira JE, Lam YW, Huston CD. Coactosin Phosphorylation Controls Entamoeba histolytica Cell Membrane Protrusions and Cell Motility. mBio 2020; 11:e00660-20. [PMID: 32753489 PMCID: PMC7407079 DOI: 10.1128/mbio.00660-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
Invasion of the colon wall by Entamoeba histolytica during amoebic dysentery entails migration of trophozoites through tissue layers that are rich in extracellular matrix. Transcriptional silencing of the E. histolytica surface metalloprotease EhMSP-1 produces hyperadherent less-motile trophozoites that are deficient in forming invadosomes. Reversible protein phosphorylation is often implicated in regulation of cell motility and invadosome formation. To identify such intermediaries of the EhMSP-1-silenced phenotype, here we compared the phosphoproteomes of EhMSP-1-silenced and vector control trophozoites by using quantitative tandem mass spectrometry-based proteomics. Six proteins were found to be differentially phosphorylated in EhMSP-1-silenced and control cells, including EhCoactosin, a member of the ADF/cofilin family of actin-binding proteins, which was more frequently phosphorylated at serine 147. Regulated overexpression of wild-type, phosphomimetic, and nonphosphorylatable EhCoactosin variants was used to test if phosphorylation functions in control of E. histolytica actin dynamics. Each of the overexpressed proteins colocalized with F-actin during E. histolytica phagocytosis. Nonetheless, trophozoites overexpressing an EhCoactosin phosphomimetic mutant formed more and poorly coordinated cell membrane protrusions compared to those in control or cells expressing a nonphosphorylatable mutant, while trophozoites overexpressing nonphosphorylatable EhCoactosin were significantly more motile within a model of mammalian extracellular matrix. Therefore, although EhCoactosin's actin-binding ability appeared unaffected by phosphorylation, EhCoactosin phosphorylation helps to regulate amoebic motility. These data help to understand the mechanisms underlying altered adherence and motility in EhMSP-1-silenced trophozoites and lay the groundwork for identifying kinases and phosphatases critical for control of amoebic invasiveness.IMPORTANCE Invasive amoebiasis, caused by the intestinal parasite Entamoeba histolytica, causes life-threatening diarrhea and liver abscesses, but, for unknown reasons, only approximately 10% of E. histolytica infections become symptomatic. A key requirement of invasion is the ability of the parasite to migrate through tissue layers. Here, we systematically looked for differences in protein phosphorylation between control parasites and a previously identified hyperadherent E. histolytica cell line that has reduced motility. We identified EhCoactosin, an actin-binding protein not previously known to be phosphoregulated, as one of the differentially phosphorylated proteins in E. histolytica and demonstrated that EhCoactosin phosphorylation functions in control of cell membrane dynamics and amoebic motility. This and the additional differentially phosphorylated proteins reported lay the groundwork for identifying kinases and phosphatases that regulate tissue invasiveness.
Collapse
Affiliation(s)
- Muhammad M Hasan
- Department of Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, USA
| | - José E Teixeira
- Department of Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Ying-Wai Lam
- Proteomics Facility, Vermont Genetics Network, University of Vermont, Burlington, Vermont, USA
- Department of Biology, University of Vermont, Burlington, Vermont, USA
| | - Christopher D Huston
- Department of Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
10
|
Pei B, Li T, Qian Q, Fan W, He X, Zhu Y, Xu L. Downregulation of microRNA-30c-5p was responsible for cell migration and tumor metastasis via COTL1-mediated microfilament arrangement in breast cancer. Gland Surg 2020; 9:747-758. [PMID: 32775265 DOI: 10.21037/gs-20-472] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Breast cancer metastasis is the main problem that affects the therapy and prognosis of breast cancer patients. Studies have indicated the role of microRNAs in breast cancer regulation, but the mechanisms are largely unknown. Methods In this study, we determined the expression of microRNA-30c-5p (miR-30c-5p) and coactosin-like protein 1 (COTL1) gene in breast cancer tissues, and revealed their effects on breast cancer metastasis regulation. Breast cancer and paracancerous tissues were collected. Reverse transcriptase polymerase chain reaction (RT-PCR) was used to analyze the expression of miR-30c-5p and COTL1, and breast cancer cell line (MCF-7) was employed to verify the relationship between miR-30c-5p and COTL1. Western blot analysis and immunofluorescence were used for proteins analysis and microfilament observation, respectively. A dual-luciferase reporter gene was used for microRNA-gene interaction assay. Results The results showed that the expression of miR-30c-5p decreased, while the expression of COTL1 increased in breast cancer tissues. The results of luciferase reporting gene assay showed that, COTL1 was the target of miR-30c-5p. After miR-30c-5p was upregulated, the expression of COTL1 was reduced, microfilament arrangement was in disorder, and cell migration ability was inhibited. After miR-30c-5p was downregulated, the expression of COTL1 was increased, and the cell migration ability was enhanced. COTL1 protein expression levels were significantly higher in cancer tissues with lymph node metastasis. Conclusions These findings indicate that miR-30c-5p/COTL1 pathway regulates breast cancer metastasis and can be used as a potential therapy target.
Collapse
Affiliation(s)
- Bei Pei
- Changzhou No. 2 People's Hospital, Xinglong Lance, Changzhou, China.,Department of Breast Surgery, Changzhou No. 2 People's Hospital, Changzhou 213003, China
| | | | - Qi Qian
- Changzhou No. 2 People's Hospital, Xinglong Lance, Changzhou, China.,Department of Breast Surgery, Changzhou No. 2 People's Hospital, Changzhou 213003, China
| | - Wenqiang Fan
- Changzhou No. 2 People's Hospital, Xinglong Lance, Changzhou, China.,Department of Breast Surgery, Changzhou No. 2 People's Hospital, Changzhou 213003, China
| | - Xiao He
- Changzhou No. 2 People's Hospital, Xinglong Lance, Changzhou, China.,Department of Breast Surgery, Changzhou No. 2 People's Hospital, Changzhou 213003, China
| | - Yulan Zhu
- Changzhou No. 2 People's Hospital, Xinglong Lance, Changzhou, China.,Department of Breast Surgery, Changzhou No. 2 People's Hospital, Changzhou 213003, China
| | - Lingyun Xu
- Changzhou No. 2 People's Hospital, Xinglong Lance, Changzhou, China.,Department of Breast Surgery, Changzhou No. 2 People's Hospital, Changzhou 213003, China.,Dalian Medical University, Dalian, China
| |
Collapse
|
11
|
Arruda B, Piñeyro P, Derscheid R, Hause B, Byers E, Dion K, Long D, Sievers C, Tangen J, Williams T, Schwartz K. PCV3-associated disease in the United States swine herd. Emerg Microbes Infect 2019; 8:684-698. [PMID: 31096848 PMCID: PMC6534263 DOI: 10.1080/22221751.2019.1613176] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Porcine circovirus-associated disease encompasses multiple disease syndromes including porcine circovirus 2 systemic diseases, reproductive failure, and porcine dermatitis and nephropathy syndrome. Until recently, porcine circovirus 2 was the only species associated with the porcine circovirus-associated disease. In this report, diagnostic investigations of thirty-six field cases submitted from multiple production systems, numerous sites and varied geographic locations demonstrated porcine circovirus 3 within lesions by in situ hybridization including fetuses with myocarditis, weak-born neonatal piglets with encephalitis and myocarditis, from cases of porcine dermatitis and nephropathy syndrome, and in weaned pigs with systemic periarteritis. Porcine circovirus 3 was detected by PCR in numerous fetuses and perinatal piglets at high viral loads (trillions of genome copies per mL of tissue homogenate). Samples from all cases in this study were assayed and found negative for porcine circovirus 2 by PCR. Metagenomic sequencing was performed on a subset of reproductive cases, consisting of sixteen fetuses/fetal sample pools. PCV3 was identified in all pools and the only virus identified in fourteen pools. Based on these data, porcine circovirus 3 is considered a putative cause of reproductive failure, encephalitis and myocarditis in perinatal piglets, porcine dermatitis and nephropathy syndrome, and periarteritis in swine in the United States.
Collapse
Affiliation(s)
- Bailey Arruda
- a Department of Veterinary Diagnostic and Production Animal Medicine , Iowa State University , Ames , IA , USA
| | - Pablo Piñeyro
- a Department of Veterinary Diagnostic and Production Animal Medicine , Iowa State University , Ames , IA , USA
| | - Rachel Derscheid
- a Department of Veterinary Diagnostic and Production Animal Medicine , Iowa State University , Ames , IA , USA
| | - Ben Hause
- b Cambridge Technologies , Worthington , MN , USA
| | | | - Kate Dion
- d The Hanor Company of Wisconsin, LLC , Enid , OK , USA
| | | | | | - Jon Tangen
- d The Hanor Company of Wisconsin, LLC , Enid , OK , USA
| | | | - Kent Schwartz
- a Department of Veterinary Diagnostic and Production Animal Medicine , Iowa State University , Ames , IA , USA
| |
Collapse
|
12
|
Further studies about Coactosin-like protein-1 affecting the migration of mouse neocortical neurons. J Mol Histol 2018; 49:519-530. [PMID: 30128637 DOI: 10.1007/s10735-018-9790-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022]
Abstract
During the development of mammalian cortex, late neurons generated by neuronal progenitors bypass earlier-born neurons and migrate to reach upper layers of cortical plate in an inner-to-outer fashion. Filamentous-actin (F-actin) can regulate neuronal migration, whereas Coactosin-like protein 1 (Cotl1) modulates F-actin. Lys 75 and Arg 73 of Cotl1 play an important role in binding F-actin; when they are mutated to Glu, Cotl1 cannot bind F-actin, called as a non-actin-binding mutant (ABM). The Lys 131 site of Cotl1, the 5-Lipoxygenase (5LO) binding site, is spatially close to Lys 75, leading to impact the binding of Cotl1 to F-actin. When Lys 131 is mutated to Ala (K131A), Cotl1 cannot bind to 5LO. We have demonstrated that overexpression of Cotl1 inhibited neuronal migration and increased the length of neuronal leading processes. To further explore cellular and molecular mechanisms of Cotl1's effect on neuronal migration, we constructed two mutant vectors-Cotl1-ABM and Cotl1-K131A and studied using in utero electroporation and primary neuronal culture technique. Results indicated that in the Cotl1-ABM group, the neuronal migration and length of the leading process both recovered as control neurons at the postnatal day 1 (P1), while in the Cotl1-K131A group, numerous neurons remained in deeper layers of cortical plate or intermediate zone. However, at P7, most Cotl1-K131A transfected neurons reached their destination. Moreover, we found that overexpression of Cotl1 inhibited the proliferation and mitotic activity of NPs. Therefore, These results demonstrated that Cotl1 played an important role in mouse neocortical development.
Collapse
|
13
|
Cheng X, Li K, Liu M, Hu X, Xu M, Yan R, Zhao S. P85 regulates neuronal migration through affecting neuronal morphology during mouse corticogenesis. Cell Tissue Res 2017; 372:23-31. [PMID: 29130119 DOI: 10.1007/s00441-017-2707-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 10/04/2017] [Indexed: 11/25/2022]
Abstract
In mammalian developing embryonic cortex, projection neurons migrate from the ventricular zone to the cortical plate, guided by radial glial cells with a transformation between bipolar and multipolar morphology. Previous studies have demonstrated that the PI3K-Akt-mTOR signal plays a critical role in brain development. However, the function of P85 in cortical development is still unclear. In the present study, we found that overexpression of P85 impaired cortical neuronal migration. Using in utero electroporation, we revealed that the length of the leading process in P85 overexpressed neurons became shorter than that in the control group but with more branches. Using markers for new-born neurons, we further found that overexpression of P85 did not affect the ultimate fate of these cortical neurons. These findings indicated that the P85 subunit plays an essential role in neuronal migration and neuronal morphology during mouse corticogenesis.
Collapse
Affiliation(s)
- Xinran Cheng
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Kaikai Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - MengMeng Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Xinde Hu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mingrui Xu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Runchuan Yan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China.
| |
Collapse
|