1
|
Li DZ, Yang ZY, Leng A, Zhang Q, Zhang XD, Bian YC, Xiao R, Ren JJ. Targeting AGTPBP1 inhibits pancreatic cancer progression via regulating microtubules and ERK signaling pathway. Mol Med 2024; 30:119. [PMID: 39129004 PMCID: PMC11318240 DOI: 10.1186/s10020-024-00892-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND AGTPBP1 is a cytosolic carboxypeptidase that cleaves poly-glutamic acids from the C terminus or side chains of α/β tubulins. Although its dysregulated expression has been linked to the development of non-small cell lung cancer, the specific roles and mechanisms of AGTPBP1 in pancreatic cancer (PC) have yet to be fully understood. In this study, we examined the role of AGTPBP1 on PC in vitro and in vivo. METHODS Immunohistochemistry was used to examine the expression of AGTPBP1 in PC and non-cancerous tissues. Additionally, we assessed the malignant behaviors of PC cells following siRNA-mediated AGTPBP1 knockdown both in vitro and in vivo. RNA sequencing and bioinformatics analysis were performed to identify the differentially expressed genes regulated by AGTPBP1. RESULTS We determined that AGTPBP1 was overexpressed in PC tissues and the higher expression of AGTPBP1 was closely related to the location of tumors. AGTPBP1 inhibition can significantly decrease cell progression in vivo and in vitro. Moreover, the knockdown of AGTPBP1 inhibited the expression of ERK1/2, P-ERK1/2, MYLK, and TUBB4B proteins via the ERK signaling pathway. CONCLUSION Our research indicates that AGTPBP1 may be a putative therapeutic target for PC.
Collapse
Affiliation(s)
- Ding-Zhong Li
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China
| | - Zhe-Yu Yang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China
| | - Asi Leng
- Medical Simulation Center, Inner Mongolia Medical University, Huhhot, 010059, PR China
| | - Qian Zhang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China
| | - Xiao-Dong Zhang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China
| | - Yan-Chao Bian
- Inner Mongolia Key Laboratory of Molecular Pathology, Inner Mongolia Medical University, #5, Xin Hua Street, Huhhot, 010059, PR China
| | - Rui Xiao
- Inner Mongolia Key Laboratory of Molecular Pathology, Inner Mongolia Medical University, #5, Xin Hua Street, Huhhot, 010059, PR China.
| | - Jian-Jun Ren
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China.
| |
Collapse
|
2
|
Wei M, Zhang N, Li XD. Characterisation of the myosin light chain kinase (MLCK) gene of Locusta migratoria and the encoded MLCK. INSECT MOLECULAR BIOLOGY 2024; 33:338-349. [PMID: 38411321 DOI: 10.1111/imb.12902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
Myosin light chain kinase (MLCK) is a dedicated kinase of myosin regulatory light chain (RLC), playing an essential role in the regulation of muscle contraction and cell motility. Much of the knowledge about MLCK comes from the study of vertebrate MLCK, and little is known about insect MLCK. Here, we identified the single MLCK gene in the locust Locusta migratoria, which spans over 1400 kb, includes 62 exons and accounts for at least five transcripts. We found that the five distinct transcripts of the locust MLCK gene are expressed in a tissue-specific manner, including three muscle-specific isoforms and two generic isoforms. To characterise the kinase activity of locust MLCK, we recombinantly expressed LmMLCK-G, the smallest locust MLCK isoform, in insect Sf9 cells. We demonstrated that LmMLCK-G is a Ca2+/calmodulin-dependent kinase that specifically phosphorylates serine 50 of locust muscle myosin RLC (LmRLC). Additionally, we found that almost all LmRLC molecules in the flight muscle and the hindleg muscles of adult locusts are phosphorylated.
Collapse
Affiliation(s)
- Miao Wei
- State Key Laboratory of Integrated Management of Insect Pests and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ning Zhang
- State Key Laboratory of Integrated Management of Insect Pests and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiang-Dong Li
- State Key Laboratory of Integrated Management of Insect Pests and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Lymperopoulos A, Borges JI, Stoicovy RA. RGS proteins and cardiovascular Angiotensin II Signaling: Novel opportunities for therapeutic targeting. Biochem Pharmacol 2023; 218:115904. [PMID: 37922976 PMCID: PMC10841918 DOI: 10.1016/j.bcp.2023.115904] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Angiotensin II (AngII), as an octapeptide hormone normally ionized at physiological pH, cannot cross cell membranes and thus, relies on, two (mainly) G protein-coupled receptor (GPCR) types, AT1R and AT2R, to exert its intracellular effects in various organ systems including the cardiovascular one. Although a lot remains to be elucidated about the signaling of the AT2R, AT1R signaling is known to be remarkably versatile, mobilizing a variety of G protein-dependent and independent signal transduction pathways inside cells to produce a biological outcome. Cardiac AT1R signaling leads to hypertrophy, adverse remodeling, fibrosis, while vascular AT1R signaling raises blood pressure via vasoconstriction, but also elicits hypertrophic, vascular growth/proliferation, and pathological remodeling sets of events. In addition, adrenal AT1R is the major physiological stimulus (alongside hyperkalemia) for secretion of aldosterone, a mineralocorticoid hormone that contributes to hypertension, electrolyte abnormalities, and to pathological remodeling of the failing heart. Regulator of G protein Signaling (RGS) proteins, discovered about 25 years ago as GTPase-activating proteins (GAPs) for the Gα subunits of heterotrimeric G proteins, play a central role in silencing G protein signaling from a plethora of GPCRs, including the AngII receptors. Given the importance of AngII and its receptors, but also of several RGS proteins, in cardiovascular homeostasis, the physiological and pathological significance of RGS protein-mediated modulation of cardiovascular AngII signaling comes as no surprise. In the present review, we provide an overview of the current literature on the involvement of RGS proteins in cardiovascular AngII signaling, by discussing their roles in cardiac (cardiomyocyte and cardiofibroblast), vascular (smooth muscle and endothelial cell), and adrenal (medulla and cortex) AngII signaling, separately. Along the way, we also highlight the therapeutic potential of enhancement of, or, in some cases, inhibition of each RGS protein involved in AngII signaling in each one of these cell types.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University Barry and Judy Silverman College of Pharmacy, Fort Lauderdale, FL 33328-2018, USA.
| | - Jordana I Borges
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University Barry and Judy Silverman College of Pharmacy, Fort Lauderdale, FL 33328-2018, USA
| | - Renee A Stoicovy
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University Barry and Judy Silverman College of Pharmacy, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
4
|
Khapchaev AY, Antonova OA, Kazakova OA, Samsonov MV, Vorotnikov AV, Shirinsky VP. Long-Term Experimental Hyperglycemia Does Not Impair Macrovascular Endothelial Barrier Integrity and Function in vitro. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1126-1138. [PMID: 37758312 DOI: 10.1134/s0006297923080072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 10/03/2023]
Abstract
Hyperglycemia is a hallmark of type 2 diabetes implicated in vascular endothelial dysfunction and cardiovascular complications. Many in vitro studies identified endothelial apoptosis as an early outcome of experimentally modeled hyperglycemia emphasizing cell demise as a significant factor of vascular injury. However, endothelial apoptosis has not been observed in vivo until the late stages of type 2 diabetes. Here, we studied the long-term (up to 4 weeks) effects of high glucose (HG, 30 mM) on human umbilical vein endothelial cells (HUVEC) in vitro. HG did not alter HUVEC monolayer morphology, ROS levels, NO production, and exerted minor effects on the HUVEC apoptosis markers. The barrier responses to various clues were indistinguishable from those by cells cultured in physiological glucose (5 mM). Tackling the key regulators of cytoskeletal contractility and endothelial barrier revealed no differences in the histamine-induced intracellular Ca2+ responses, nor in phosphorylation of myosin regulatory light chain or myosin light chain phosphatase. Altogether, these findings suggest that vascular endothelial cells may well tolerate HG for relatively long exposures and warrant further studies to explore mechanisms involved in vascular damage in advanced type 2 diabetes.
Collapse
Affiliation(s)
- Asker Y Khapchaev
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia.
| | - Olga A Antonova
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Olga A Kazakova
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Mikhail V Samsonov
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Alexander V Vorotnikov
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Vladimir P Shirinsky
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| |
Collapse
|
5
|
Identification of AGR2 Gene-Specific Expression Patterns Associated with Epithelial-Mesenchymal Transition. Int J Mol Sci 2022; 23:ijms231810845. [PMID: 36142758 PMCID: PMC9504245 DOI: 10.3390/ijms231810845] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022] Open
Abstract
The TGF-β signaling pathway is involved in numerous cellular processes, and its deregulation may result in cancer development. One of the key processes in tumor progression and metastasis is epithelial to mesenchymal transition (EMT), in which TGF-β signaling plays important roles. Recently, AGR2 was identified as a crucial component of the cellular machinery responsible for maintaining the epithelial phenotype, thereby interfering with the induction of mesenchymal phenotype cells by TGF-β effects in cancer. Here, we performed transcriptomic profiling of A549 lung cancer cells with CRISPR-Cas9 mediated AGR2 knockout with and without TGF-β treatment. We identified significant changes in transcripts associated with focal adhesion and eicosanoid production, in particular arachidonic acid metabolism. Changes in transcripts associated with the focal adhesion pathway were validated by RT-qPCR of COL4A1, COL4A2, FLNA, VAV3, VEGFA, and VINC mRNAs. In addition, immunofluorescence showed the formation of stress fibers and vinculin foci in cells without AGR2 and in response to TGF-β treatment, with synergistic effects observed. These findings imply that both AGR2 downregulation and TGF-β have a role in focal adhesion formation and cancer cell migration and invasion. Transcripts associated with arachidonic acid metabolism were downregulated after both AGR2 knockout and TGF-β treatment and were validated by RT-qPCR of GPX2, PTGS2, and PLA2G4A. Since PGE2 is a product of arachidonic acid metabolism, its lowered concentration in media from AGR2-knockout cells was confirmed by ELISA. Together, our results demonstrate that AGR2 downregulation and TGF-β have an essential role in focal adhesion formation; moreover, we have identified AGR2 as an important component of the arachidonic acid metabolic pathway.
Collapse
|
6
|
Shan H, Ren K, Liu J, Rehman SU, Yan X, Ma X, Zheng Y, Feng T, Wang X, Li Z, Zhou W, Chuang C, Liang M, Zheng J, Liu Q. Comprehensive Transcriptome Sequencing Analysis of Hirudinaria manillensis in Different Growth Periods. Front Physiol 2022; 13:897458. [PMID: 35694407 PMCID: PMC9174698 DOI: 10.3389/fphys.2022.897458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
Medical leeches are widely been used in biochemical and clinical medical studies, helping to restore blood circulation to grafted or severely injured tissue. Mostly, adult leeches are being used in the traditional pharmacopeia, but the gene expression profiling of leeches in different growth periods is not well-reported. So, in this study, we used transcriptome analysis to analyze the comparative gene expression patterns of Hirudinaria manillensis (H. manillensis) in different growth periods, including larval, young, and adult stages. We constructed 24 cDNA libraries from H. manillensis larval, young, and adult stages, and about 54,639,118 sequences were generated, 18,106 mRNA transcripts of which 958 novel mRNAs and 491 lncRNAs were also assembled as well. Furthermore, the results of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that the differentially upregulated genes from the larval to adult stages were enriched in pathways such as cilium, myofibril, contractile fiber, cytoskeleton proteins, dilated cardiomyopathy, adrenergic signaling in cardiomyocytes, etc. Moreover, in the adult stages, a significant increase in the expression of the Hirudin-HM (HIRM2) genes was detected. In addition, our comparative transcriptome profiling data from different growth stages of H. manillensis also identified a large number of DEGs and DElncRNAs which were tentatively found to be associated with the growth of H. manillensis; as it grew, the muscle-related gene expression increased, while the lipid metabolism and need for stimulation and nutrition-related genes decreased. Similarly, the higher expression of HIRM2 might attribute to the high expression of protein disulfide isomerase gene family (PDI) family genes in adulthood, which provides an important clue that why adult leeches rather than young leeches are widely used in clinical therapeutics and traditional Chinese medicine.
Collapse
Affiliation(s)
- Huiquan Shan
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Ke Ren
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jiasheng Liu
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Saif ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Xiuying Yan
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Xiaocong Ma
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Yalin Zheng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Tong Feng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Xiaobo Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Weiguan Zhou
- THAI Natural Hirudin Co., Ltd., Bangkok, Thailand
| | - Chen Chuang
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mingkun Liang
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jinghui Zheng
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
- *Correspondence: Jinghui Zheng, ; Qingyou Liu,
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
- *Correspondence: Jinghui Zheng, ; Qingyou Liu,
| |
Collapse
|
7
|
Li M, Yang X, Masoudi A, Xiao Q, Li N, Wang N, Chang G, Ren S, Li H, Liu J, Wang H. The regulatory strategy of proteins in the mouse kidney during Babesia microti infection. Exp Parasitol 2022; 235:108232. [DOI: 10.1016/j.exppara.2022.108232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/03/2022] [Accepted: 02/10/2022] [Indexed: 11/04/2022]
|
8
|
Shi X, Yu X, Wang J, Bian S, Li Q, Fu F, Zou X, Zhang L, Bast RC, Lu Z, Guo L, Chen Y, Zhou J. SIK2 promotes ovarian cancer cell motility and metastasis by phosphorylating MYLK. Mol Oncol 2022; 16:2558-2574. [PMID: 35278271 PMCID: PMC9251837 DOI: 10.1002/1878-0261.13208] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/11/2022] [Accepted: 03/10/2022] [Indexed: 11/10/2022] Open
Abstract
Salt‐inducible kinase 2 (SIK2; also known as serine/threonine‐protein kinase SIK2) is overexpressed in several cancers and has been implicated in cancer progression. However, the mechanisms by which SIK2 regulates cancer cell motility, migration and metastasis in ovarian cancer have not been fully discovered. Here, we identify that SIK2 promotes ovarian cancer cell motility, migration and metastasis in vitro and in vivo. Mechanistically, SIK2 regulated cancer cell motility and migration by myosin light chain kinase, smooth muscle (MYLK)‐meditated phosphorylation of myosin light chain 2 (MYL2). SIK2 directly phosphorylated MYLK at Ser343 and activated its downstream effector MYL2, promoting ovarian cancer cell motility and metastasis. In addition, we found that adipocytes induced SIK2 phosphorylation at Ser358 and MYLK phosphorylation at Ser343, enhancing ovarian cancer cell motility. Moreover, SIK2 protein expression was positively correlated with the expression of MYLK‐pS343 in ovarian cancer cell lines and tissues. The co‐expression of SIK2 and MYLK‐pS343 was associated with reduced median overall survival in human ovarian cancer samples. Taken together, SIK2 positively regulates ovarian cancer motility, migration and metastasis, suggesting that SIK2 is a potential candidate for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xiu Shi
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
- Jiangsu Institute of Clinical Immunology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Xuejiao Yu
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Juan Wang
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Shimin Bian
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Qiutong Li
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Fengqing Fu
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
- Jiangsu Institute of Clinical Immunology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Xinwei Zou
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Lin Zhang
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Robert C. Bast
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Zhen Lu
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Lingchuan Guo
- Department of Experimental Therapeutics University of Texas M.D. Anderson Cancer Center Houston Texas USA
| | - Youguo Chen
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
| |
Collapse
|
9
|
Anandita NW, Nurdiana N, Wahyuni ES, Sujuti H. Cyclic Adenosine Monophosphate, Inositol 1,4,5-trisphosphate, Calcium, and Phosphorylated Myosin Light Chain Regulation Through M2 and M3 Muscarinic Receptors of Scleral Fibroblast Cells in Rat Myopia Model. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIM: This study aims to investigate the concentration of cyclic adenosine monophosphate (cAMP), inositol 1,4,5-trisphosphate (IP3), calcium (Ca2+), and the expression phosphorylated myosin light chain (MLC) in Rattus norvegicus scleral fibroblast cells.
METHODOLOGY: This study utilized an in vitro experimental study by applying Rattus norvegicus scleral fibroblast cell culture. The cultured cells were divided into control and lens-induced myopia (LIM) groups. The control and LIM culture groups were each divided into five groups, namely, negative control, 0.1 μM acetylcholine, 0.1 μM himbacine, 0.1 μM methoctramine, and 0.1 μM 4-DAMP group. The cAMP, IP3, and Ca2+ concentration were analyzed in the 0th, 5th, 10th, 20th, and 30th. The phosphorylated MLC expression was analyzed using confocal microscope.
RESULTS: In the LIM group, the highest cAMP concentration is visible at the 10th min on the himbacine group (0.304 ± 0; p = 0.043) and on the 4-DAMP group (0.346 ± 0; p = 0.043). The highest IP3 concentration is found on the LIM group at the 20th min in comparison to the control group (2503.6 ± 11 vs. 2039.2 ± 2.1; p = 0.046). The highest Ca2+ concentration belongs to the 4-DAMP treatment group from the 5th to the 30th min. The highest average phosphorylated MLC expression value in the LIM group is shown by the 0.1μM 4-DAMP treatment (184.2 ± 37.9c au).
CONCLUSION: The regulation of cAMP, IP3, Ca2+, and phosphorylated MLC on the M2 and M3 muscarinic receptor of the scleral fibroblast cells of myopia animal models differs from normal animal models which may be due to interactions of M2 and M3 muscarinic receptor as compensation reaction or crosstalk on myopia induction.
Collapse
|
10
|
Sorensen DW, Injeti ER, Mejia-Aguilar L, Williams JM, Pearce WJ. Postnatal development alters functional compartmentalization of myosin light chain kinase in ovine carotid arteries. Am J Physiol Regul Integr Comp Physiol 2021; 321:R441-R453. [PMID: 34318702 PMCID: PMC8530762 DOI: 10.1152/ajpregu.00293.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The rate-limiting enzyme for vascular contraction, myosin light chain kinase (MLCK), phosphorylates regulatory myosin light chain (MLC20) at rates that appear faster despite lower MLCK abundance in fetal compared with adult arteries. This study explores the hypothesis that greater apparent tissue activity of MLCK in fetal arteries is due to age-dependent differences in intracellular distribution of MLCK in relation to MLC20. Under optimal conditions, common carotid artery homogenates from nonpregnant adult female sheep and near-term fetuses exhibited similar values of Vmax and Km for MLCK. A custom-designed, computer-controlled apparatus enabled electrical stimulation and high-speed freezing of arterial segments at exactly 0, 1, 2, and 3 s, calculation of in situ rates of MLC20 phosphorylation, and measurement of time-dependent colocalization between MLCK and MLC20. The in situ rate of MLC20 phosphorylation divided by total MLCK abundance averaged to values 147% greater in fetal (1.06 ± 0.28) than adult (0.43 ± 0.08) arteries, which corresponded, respectively, to 43 ± 10% and 31 ± 3% of the Vmax values measured in homogenates. Confocal colocalization analysis revealed in fetal and adult arteries that 33 ± 6% and 20 ± 5% of total MLCK colocalized with pMLC20, and that MLCK activation was greater in periluminal than periadventitial regions over the time course of electrical stimulation in both age groups. Together, these results demonstrate that the catalytic activity of MLCK is similar in fetal and adult arteries, but that the fraction of total MLCK in the functional compartment involved in contraction is significantly greater in fetal than adult arteries.
Collapse
Affiliation(s)
- Dane W Sorensen
- Division of Physiology, Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Elisha R Injeti
- Department of Pharmaceutical Sciences, Cedarville University School of Pharmacy, Cedarville, Ohio
| | - Luisa Mejia-Aguilar
- Division of Physiology, Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - James M Williams
- Division of Physiology, Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - William J Pearce
- Division of Physiology, Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
11
|
Khapchaev AY, Watterson DM, Shirinsky VP. Phosphorylation-dependent subcellular redistribution of small myosin light chain kinase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119104. [PMID: 34302892 DOI: 10.1016/j.bbamcr.2021.119104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Myosin light chain kinase (MLCK) is a Ca2+-calmodulin-dependent enzyme dedicated to phosphorylate and activate myosin II to provide force for various motile processes. In smooth muscle cells and many other cells, small MLCK (S-MLCK) is a major isoform. S-MLCK is an actomyosin-binding protein firmly attached to contractile machinery in smooth muscle cells. Still, it can leave this location and contribute to other cellular processes. However, molecular mechanisms for switching the S-MLCK subcellular localization have not been described. METHODS Site-directed mutagenesis and in vitro protein phosphorylation were used to study functional roles of discrete in-vivo phosphorylated residues within the S-MLCK actin-binding domain. In vitro co-sedimentation analysis was applied to study the interaction of recombinant S-MLCK actin-binding fragment with filamentous actin. Subcellular distribution of phosphomimicking S-MLCK mutants was studied by fluorescent microscopy and differential cell extraction. RESULTS Phosphorylation of S-MLCK actin-binding domain at Ser25 and/or Thr56 by proline-directed protein kinases or phosphomimicking these posttranslational modifications alters S-MLCK binding to actin filaments both in vitro and in cells, and induces S-MLCK subcellular translocation with no effect on the enzyme catalytic properties. CONCLUSIONS Phosphorylation of the amino terminal actin-binding domain of S-MLCK renders differential subcellular targeting of the enzyme and may, thereby, contribute to a variety of context-dependent responses of S-MLCK to cellular and tissue stimuli. GENERAL SIGNIFICANCE S-MLCK physiological function can potentially be modulated via phosphorylation of its actin recognition domain, a regulation distinct from the catalytic and calmodulin regulatory domains.
Collapse
Affiliation(s)
- Asker Y Khapchaev
- National Medical Research Center for Cardiology, 3rd Cherepkovskaya St., 15a, Moscow 121552, Russian Federation.
| | | | - Vladimir P Shirinsky
- National Medical Research Center for Cardiology, 3rd Cherepkovskaya St., 15a, Moscow 121552, Russian Federation
| |
Collapse
|
12
|
Chen X, Hu C, Fan X, Wang Y, Li Q, Su YQ, Zhang DM, Yang Q, Passerini AG, Sun C. mTOR Inhibition Promotes Pneumonitis Through Inducing Endothelial Contraction and Hyperpermeability. Am J Respir Cell Mol Biol 2021; 65:646-657. [PMID: 34251297 DOI: 10.1165/rcmb.2020-0390oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Compromised endothelial (EC) barrier function is a hallmark of inflammatory diseases. Mammalian target of rapamycin (mTOR) inhibitors, widely applied as clinical therapies, cause pneumonitis through mechanisms not yet fully understood. This study aimed to elucidate the EC mechanisms underlying the pathogenesis of pneumonitis caused by mTOR inhibition (mTORi). Mice with EC-specific deletion of mTOR complex components (Mtor, Rptor or Rictor) were administered LPS to induce pulmonary injury. Cultured EC were treated with pharmacological inhibitors, small interfering RNA or overexpression-plasmids. EC barrier function was evaluated in vivo with Evan's blue assay and in vitro by measurement of transendothelial electrical resistance and albumin flux. mTORi increased basal and TNFα-induced EC permeability, which was caused by myosin light chain (MLC) phosphorylation-dependent cell contraction. Inactivation of mTOR kinase activity by mTORi triggered PKCδ/p38/NF-κB signaling that significantly upregulated TNFα-induced MLC kinase (MLCK) expression, while Raptor promoted the phosphorylation of PKCα/MYPT1 independent of its interaction with mTOR, leading to suppression of MLC phosphatase (MLCP) activity. EC-specific deficiency in mTOR, Raptor or Rictor aggravated lung inflammation in LPS-treated mice. These findings reveal that mTORi induces PKC-dependent endothelial MLC phosphorylation, contraction and hyperpermeability that promote pneumonitis.
Collapse
Affiliation(s)
- Xiaolin Chen
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,2Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Chengxiu Hu
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Xing Fan
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Yiying Wang
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Qiannan Li
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - You-Qiang Su
- Nanjing Medical University, 12461, State Key Laboratory of Reproductive Medicine, Nanjing, China
| | - Dai-Min Zhang
- Nanjing First Hospital, Nanjing Medical University, Department of Cardiology, Nanjing, China
| | - QianLu Yang
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Anthony G Passerini
- University of California Davis, 8789, Department of Biomedical Engineering, Davis, California, United States
| | - ChongXiu Sun
- Nanjing Medical University, 12461, Nanjing, China;
| |
Collapse
|
13
|
Shiravand Y, Walter U, Jurk K. Fine-Tuning of Platelet Responses by Serine/Threonine Protein Kinases and Phosphatases-Just the Beginning. Hamostaseologie 2021; 41:206-216. [PMID: 34192779 DOI: 10.1055/a-1476-7873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Comprehensive proteomic analyses of human and murine platelets established an extraordinary intracellular repertoire of signaling components, which control crucial functions. The spectrum of platelet serine/threonine protein kinases (more than 100) includes the AGC family (protein kinase A, G, C [PKA, PKG, PKC]), the mitogen-activated protein kinases (MAPKs), and others. PKA and PKG have multiple significantly overlapping substrates in human platelets, which possibly affect functions with clear "signaling nodes" of regulation by multiple protein kinases/phosphatases. Signaling nodes are intracellular Ca2+ stores, the contractile system (myosin light chains), and other signaling components such as G-proteins, protein kinases, and protein phosphatases. An example for this fine-tuning is the tyrosine kinase Syk, a crucial component of platelet activation, which is controlled by several serine/threonine and tyrosine protein kinases as well as phosphatases. Other protein kinases including PKA/PKG modulate protein phosphatase 2A, which may be a master regulator of MAPK signaling in human platelets. Protein kinases and in particular MAPKs are targeted by an increasing number of clinically used inhibitors. However, the precise regulation and fine-tuning of these protein kinases and their effects on other signaling components in platelets are only superficially understood-just the beginning. However, promising future approaches are in sight.
Collapse
Affiliation(s)
- Yavar Shiravand
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
14
|
Berthenet K, Castillo Ferrer C, Fanfone D, Popgeorgiev N, Neves D, Bertolino P, Gibert B, Hernandez-Vargas H, Ichim G. Failed Apoptosis Enhances Melanoma Cancer Cell Aggressiveness. Cell Rep 2021; 31:107731. [PMID: 32521256 DOI: 10.1016/j.celrep.2020.107731] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 04/13/2020] [Accepted: 05/14/2020] [Indexed: 12/22/2022] Open
Abstract
Triggering apoptosis remains an efficient strategy to treat cancer. However, apoptosis is no longer a final destination since cancer cells can undergo partial apoptosis without dying. Recent evidence shows that partial mitochondrial permeabilization and non-lethal caspase activation occur under certain circumstances, although it remains unclear how failed apoptosis affects cancer cells. Using a cancer cell model to trigger non-lethal caspase activation, we find that melanoma cancer cells undergoing failed apoptosis have a particular transcriptomic signature associated with focal adhesions, transendothelial migration, and modifications of the actin cytoskeleton. In line with this, cancer cells surviving apoptosis gain migration and invasion properties in vitro and in vivo. We further demonstrate that failed apoptosis-associated gain in invasiveness is regulated by the c-Jun N-terminal kinase (JNK) pathway, whereas its RNA sequencing signature is found in metastatic melanoma. These findings advance our understanding of how cell death can both cure and promote cancer.
Collapse
Affiliation(s)
- Kevin Berthenet
- Cancer Research Center of Lyon (CRCL), INSERM 1052, CNRS 5286, Lyon, France; Cancer Cell Death Laboratory, Part of LabEx DEVweCAN, Université de Lyon, Lyon, France
| | - Camila Castillo Ferrer
- Cancer Target and Experimental Therapeutics, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France; EPHE, PSL Research University, Paris, France
| | - Deborah Fanfone
- Cancer Research Center of Lyon (CRCL), INSERM 1052, CNRS 5286, Lyon, France; Cancer Cell Death Laboratory, Part of LabEx DEVweCAN, Université de Lyon, Lyon, France
| | | | | | - Philippe Bertolino
- Cancer Research Center of Lyon (CRCL), INSERM 1052, CNRS 5286, Lyon, France
| | - Benjamin Gibert
- Cancer Research Center of Lyon (CRCL), INSERM 1052, CNRS 5286, Lyon, France; Apoptosis, Cancer and Development Laboratory, Labeled by "La Ligue Contre le Cancer," Part of LabEx DEVweCAN and Convergence PLAsCAN Institute, Lyon, France
| | - Hector Hernandez-Vargas
- Cancer Research Center of Lyon (CRCL), INSERM 1052, CNRS 5286, Lyon, France; Université Claude Bernard Lyon 1, Lyon, France
| | - Gabriel Ichim
- Cancer Research Center of Lyon (CRCL), INSERM 1052, CNRS 5286, Lyon, France; Cancer Cell Death Laboratory, Part of LabEx DEVweCAN, Université de Lyon, Lyon, France.
| |
Collapse
|
15
|
Brunner J, Ragupathy S, Borchard G. Target specific tight junction modulators. Adv Drug Deliv Rev 2021; 171:266-288. [PMID: 33617902 DOI: 10.1016/j.addr.2021.02.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Intercellular tight junctions represent a formidable barrier against paracellular drug absorption at epithelia (e.g., nasal, intestinal) and the endothelium (e.g., blood-brain barrier). In order to enhance paracellular transport of drugs and increase their bioavailability and organ deposition, active excipients modulating tight junctions have been applied. First-generation of permeation enhancers (PEs) acted by unspecific interactions, while recently developed PEs address specific physiological mechanisms. Such target specific tight junction modulators (TJMs) have the advantage of a defined specific mechanism of action. To date, merely a few of these novel active excipients has entered into clinical trials, as their lack in safety and efficiency in vivo often impedes their commercialisation. A stronger focus on the development of such active excipients would result in an economic and therapeutic improvement of current and future drugs.
Collapse
Affiliation(s)
- Joël Brunner
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Sakthikumar Ragupathy
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
16
|
Peng JL, Wu JZ, Li GJ, Wu JL, Xi YM, Li XQ, Wang L. Identification of potential biomarkers of peripheral blood mononuclear cell in hepatocellular carcinoma using bioinformatic analysis: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e24172. [PMID: 33466191 PMCID: PMC7808450 DOI: 10.1097/md.0000000000024172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 12/11/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the cause of an overwhelming number of cancer-related deaths across the world. Developing precise and noninvasive biomarkers is critical for diagnosing HCC. Our research was designed to explore potentially useful biomarkers of host peripheral blood mononuclear cell (PBMC) in HCC by integrating comprehensive bioinformatic analysis. METHODS Gene expression data of PBMC in both healthy individuals and patients with HCC were extracted from the Gene Expression Omnibus (GEO) to identify differentially expressed genes (DEGs). The gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were applied to annotate the function of DEGs. Protein-protein interaction analysis was performed to screen the hub genes from DEGs. cBioportal database analysis was performed to assess the prognostic significance of hub genes. The Cancer Cell Line Encyclopedia (CCLE) and The Human Protein Atlas (HPA) database analyses were performed to confirm the expression levels of the hub genes in HCC cells and tissue. RESULTS A total of 95 DEGs were screened. Results of the GO analysis revealed that DEGs were primarily involved in platelet degranulation, cytoplasm, and protein binding. Results of the KEGG analysis indicated that DEGs were primarily enriched in focal adhesion. Five genes, namely, myosin light chain kinase (MYLK), interleukin 1 beta (IL1B), phospholipase D1 (PLD1), cortactin (CTTN), and moesin (MSN), were identified as hub genes. A search in the CCLE and HPA database showed that the expression levels of these hub genes were remarkably increased in the HCC samples. Survival analysis revealed that the overexpression of MYLK, IL1B, and PLD1 may have a significant effect on HCC survival. The aberrant high expression levels of MYLK, IL1B, and PLD1 strongly indicated worse prognosis in patients with HCC. CONCLUSIONS The identified hub genes may be closely linked with HCC tumorigenicity and may act as potentially useful biomarkers for the prognostic prediction of HCC in PBMC samples.
Collapse
Affiliation(s)
- Jin-lin Peng
- Department of Infectious Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, Nanning, Guangxi
| | - Ji-zhou Wu
- Department of Infectious Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, Nanning, Guangxi
| | - Guo-jian Li
- Department of Infectious Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, Nanning, Guangxi
| | - Jian-lin Wu
- Department of Infectious Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, Nanning, Guangxi
| | - Yu-mei Xi
- Department of Infectious Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, Nanning, Guangxi
| | - Xiao-qing Li
- Department of Infectious Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, Nanning, Guangxi
| | - Lei Wang
- College of Health and Rehabilitation, Chengdu University of Chinese Medicine, Chengdu, Sichuan, PR China
| |
Collapse
|
17
|
Sorensen DW, Carreon D, Williams JM, Pearce WJ. Hypoxic modulation of fetal vascular MLCK abundance, localization, and function. Am J Physiol Regul Integr Comp Physiol 2021; 320:R1-R18. [PMID: 33112654 PMCID: PMC7847055 DOI: 10.1152/ajpregu.00212.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 12/26/2022]
Abstract
Changes in vascular contractility are among the most important physiological effects of acute and chronic fetal hypoxia. Given the essential role of myosin light-chain kinase (MLCK) in smooth muscle contractility and its heterogeneous distribution, this study explores the hypothesis that subcellular changes in MLCK distribution contribute to hypoxic modulation of fetal carotid artery contractility. Relative to common carotid arteries from normoxic term fetal lambs (FN), carotids from fetal lambs gestated at high altitude (3,802 m) (FH) exhibited depressed contractility without changes in MLCK mRNA or protein abundance. Patterns of confocal colocalization of MLCK with α-actin and 20-kDa regulatory myosin light chain (MLC20) enabled calculation of subcellular MLCK fractions: 1) colocalized with the contractile apparatus, 2) colocalized with α-actin distant from the contractile apparatus, and 3) not colocalized with α-actin. Chronic hypoxia did not affect MLCK abundance in the contractile fraction, despite a concurrent decrease in contractility. Organ culture for 72 h under 1% O2 decreased total MLCK abundance in FN and FH carotid arteries, but decreased the contractile MLCK abundance only in FH carotid arteries. Correspondingly, culture under 1% O2 depressed contractility more in FH than FN carotid arteries. In addition, hypoxia appeared to attenuate ubiquitin-independent proteasomal degradation of MLCK, as reported for other proteins. In aggregate, these results demonstrate that the combination of chronic hypoxia followed by hypoxic culture can induce MLCK translocation among at least three subcellular fractions with possible influences on contractility, indicating that changes in MLCK distribution are a significant component of fetal vascular responses to hypoxia.
Collapse
Affiliation(s)
- Dane W Sorensen
- Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Desirelys Carreon
- Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - James M Williams
- Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - William J Pearce
- Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
18
|
Xu F, Zhang P, Yuan M, Yang X, Chong T. Bioinformatic screening and identification of downregulated hub genes in adrenocortical carcinoma. Exp Ther Med 2020; 20:2730-2742. [PMID: 32765768 PMCID: PMC7401943 DOI: 10.3892/etm.2020.8987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
The molecular mechanisms of adrenocortical carcinoma (ACC) carcinogenesis and progression remain unclear. In the present study, three microarray datasets from the Gene Expression Omnibus database were screened, which identified a total of 96 differentially expressed genes (DEGs). A protein-protein interaction network (PPI) was established for these DEGs and module analysis was performed using STRING and Cytoscape. A total of eight hub genes were identified from the most significant module; namely, calponin 1 (CNN1), myosin light chain kinase (MYLK), cysteine and glycine rich protein 1 (CSRP1), myosin heavy chain 11 (MYH11), fibulin extracellular matrix protein 2 (EFEMP2), fibulin 1 (FBLN1), microfibril associated protein 4 (MFAP4) and fibulin 5 (FBLN5). The biological functions of these hub genes were analyzed using the DAVID online tool. Changes in the expression of hub genes did not affect overall survival; however, downregulated EFEMP2 decreased disease-free survival. CSRP1 and MFAP4 expression levels were associated with adverse clinicopathological features. In conclusion, although all eight hub genes were downregulated in ACC, they appeared to have important functions in ACC carcinogenesis and progression. Identification of these genes complements the genetic expression profile of ACC and provides insight for the diagnosis, treatment and prognosis of ACC.
Collapse
Affiliation(s)
- Fangshi Xu
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710000, P.R. China.,Department of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Zhang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710000, P.R. China
| | - Miao Yuan
- Department of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaojie Yang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710000, P.R. China
| | - Tie Chong
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710000, P.R. China
| |
Collapse
|
19
|
Grogan A, Tsakiroglou P, Kontrogianni-Konstantopoulos A. Double the trouble: giant proteins with dual kinase activity in the heart. Biophys Rev 2020; 12:1019-1029. [PMID: 32638332 DOI: 10.1007/s12551-020-00715-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Obscurin and its homolog, striated muscle preferentially expressed gene (SPEG), constitute a unique group of proteins abundantly expressed in striated muscles that contain two tandemly arranged MLCK-like kinases. The physiological significance of the dual kinase motifs is largely understudied; however, a collection of recent studies characterizing their binding interactions, putative targets, and disease-linked mutations have begun to shed light on their potential roles in muscle pathophysiology. Specifically, obscurin kinase 1 is proposed to regulate cardiomyocyte adhesion via phosphorylating N-cadherin, whereas SPEG kinases 1 and 2 regulate Ca2+ cycling by phosphorylating junctophilin-2 and the sarcoendoplasmic Ca2+ ATPase 2 (SERCA2). Herein, we review what is currently known regarding the potential substrates, physiological roles, and disease associations of obscurin and SPEG tandem kinase domains and provide future directions that have yet to be investigated.
Collapse
Affiliation(s)
- Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD, 21201, USA
| | - Panagiotis Tsakiroglou
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD, 21201, USA
| | | |
Collapse
|
20
|
MLCK and ROCK mutualism in endothelial barrier dysfunction. Biochimie 2020; 168:83-91. [DOI: 10.1016/j.biochi.2019.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/22/2019] [Indexed: 01/15/2023]
|
21
|
Hu X, You L, Hu C, Wu J, Ai M, He X, Huang W, Wu Z. Effects of β‑hydroxybutyric acid and ghrelin on the motility and inflammation of gastric antral smooth muscle cells involving the regulation of growth hormone secretagogue receptor. Mol Med Rep 2019; 20:5050-5058. [PMID: 31638214 PMCID: PMC6854601 DOI: 10.3892/mmr.2019.10739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 07/03/2019] [Indexed: 11/06/2022] Open
Abstract
Ghrelin is an orexigenic hormone that is produced by gastric cells. Ghrelin stimulates food intake and increases gastric movement. In rat model, injected β‑hydroxybutyric acid (β‑HB) leads to a decrease in body weight. It has been reported that patients with gastric erosions are slower to evacuate the stomach. The aim of the present study was to investigate the effects of ghrelin and β‑HB on motility and inflammation in rat gastric antral smooth muscle cells (GASMCs). GASMCs were extracted from rat gastric antrum. Cell viability was determined using the Cell Counting Kit‑8 assay. A reactive oxygen species (ROS) assay kit was used to analyze the levels of ROS using flow cytometry. Protein levels were determined using western blotting, and the expression levels of mRNAs were evaluated using reverse transcription‑quantitative PCR. β‑HB inhibited the expression of myosin regulatory light polypeptide 9 (MYL9), myosin light chain kinase (MLCK), transforming protein RhoA (RhoA), Rho‑associated protein kinase‑1 (ROCK‑1) and growth hormone secretagogue receptor (GHS‑R). By contrast, ghrelin increased the expression of MYL9, MLCK, RhoA, ROCK‑1 and GHS‑R in β‑HB‑treated GASMCs. β‑HB increased the levels of tumor necrosis factor (TNF)‑α, interleukin (IL)‑6 and ROS, and decreased the levels of manganese (Mn) superoxide dismutase (SOD), copper/zinc (Cu/Zn)SOD and catalase. Ghrelin decreased the expression of TNF‑α, IL‑6, ROS and catalase, whereas ghrelin promoted the expression of MnSOD and Cu/ZnSOD in β‑HB‑treated GASMCs. Short interfering RNA targeting GHS‑R inhibited the expression of MYL9, MLCK, RhoA and ROCK‑1, and increased the levels of TNF‑α, IL‑6 and ROS in β‑HB‑treated or ghrelin‑treated GASMCs. The present study provided preliminary evidence that β‑HB inhibits the motility of GASMCs and promotes inflammation in GASMCs, whereas ghrelin decreases these effects. GHS‑R acted as a primary regulator of motility and inflammation in GASMCs treated with β‑HB and ghrelin.
Collapse
Affiliation(s)
- Xiaolin Hu
- Department of Internal Medicine, Southwest University Hospital, Chongqing 400715, P.R. China
| | - Li You
- Department of Pharmacy, Southwest University Hospital, Chongqing 400715, P.R. China
| | - Changhua Hu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Juan Wu
- Department of Internal Medicine, Southwest University Hospital, Chongqing 400715, P.R. China
| | - Min Ai
- Department of Internal Medicine, Southwest University Hospital, Chongqing 400715, P.R. China
| | - Xiaoyan He
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Wenjie Huang
- Department of Public Health, Southwest University Hospital, Chongqing 400715, P.R. China
| | - Zonghui Wu
- Health Management Center, Southwest University Hospital, Chongqing 400715, P.R. China
| |
Collapse
|
22
|
Villalobo A, González-Muñoz M, Berchtold MW. Proteins with calmodulin-like domains: structures and functional roles. Cell Mol Life Sci 2019; 76:2299-2328. [PMID: 30877334 PMCID: PMC11105222 DOI: 10.1007/s00018-019-03062-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 02/26/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022]
Abstract
The appearance of modular proteins is a widespread phenomenon during the evolution of proteins. The combinatorial arrangement of different functional and/or structural domains within a single polypeptide chain yields a wide variety of activities and regulatory properties to the modular proteins. In this review, we will discuss proteins, that in addition to their catalytic, transport, structure, localization or adaptor functions, also have segments resembling the helix-loop-helix EF-hand motifs found in Ca2+-binding proteins, such as calmodulin (CaM). These segments are denoted CaM-like domains (CaM-LDs) and play a regulatory role, making these CaM-like proteins sensitive to Ca2+ transients within the cell, and hence are able to transduce the Ca2+ signal leading to specific cellular responses. Importantly, this arrangement allows to this group of proteins direct regulation independent of other Ca2+-sensitive sensor/transducer proteins, such as CaM. In addition, this review also covers CaM-binding proteins, in which their CaM-binding site (CBS), in the absence of CaM, is proposed to interact with other segments of the same protein denoted CaM-like binding site (CLBS). CLBS are important regulatory motifs, acting either by keeping these CaM-binding proteins inactive in the absence of CaM, enhancing the stability of protein complexes and/or facilitating their dimerization via CBS/CLBS interaction. The existence of proteins containing CaM-LDs or CLBSs substantially adds to the enormous versatility and complexity of Ca2+/CaM signaling.
Collapse
Affiliation(s)
- Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Arturo Duperier 4, 28029, Madrid, Spain.
- Instituto de Investigaciones Sanitarias, Hospital Universitario La Paz, Edificio IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.
| | - María González-Muñoz
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Arturo Duperier 4, 28029, Madrid, Spain
| | - Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, 2100, Copenhagen, Denmark.
| |
Collapse
|
23
|
Braun DJ, Bachstetter AD, Sudduth TL, Wilcock DM, Watterson DM, Van Eldik LJ. Genetic knockout of myosin light chain kinase (MLCK210) prevents cerebral microhemorrhages and attenuates neuroinflammation in a mouse model of vascular cognitive impairment and dementia. GeroScience 2019; 41:671-679. [PMID: 31104189 PMCID: PMC6885026 DOI: 10.1007/s11357-019-00072-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022] Open
Abstract
The blood-brain barrier (BBB) is critical in maintenance of brain homeostasis, and loss of its functional integrity is a key feature across a broad range of neurological insults. This includes both acute injuries such as traumatic brain injury and stroke, as well as more chronic pathologies associated with aging, such as vascular cognitive impairment and dementia (VCID). A specific form of myosin light chain kinase (MLCK210) is a major regulator of barrier integrity in general, including the BBB. Studies have demonstrated the potential of MLCK210 as a therapeutic target for peripheral disorders involving tissue barrier dysfunction, but less is known about its potential as a target for chronic neurologic disorders. We report here that genetic knockout (KO) of MLCK210 protects against cerebral microhemorrhages and neuroinflammation induced by chronic dietary hyperhomocysteinemia. Overall, the results are consistent with an accumulating body of evidence supporting MLCK210 as a potential therapeutic target for tissue barrier dysfunction and specifically implicate it in BBB dysfunction and neuroinflammation in a model of VCID.
Collapse
Affiliation(s)
- David J Braun
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Tiffany L Sudduth
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY, 40536, USA
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY, 40536, USA
| | - D Martin Watterson
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA.
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA.
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
24
|
Kazakova OA, Khapchaev AY, Ragimov AA, Salimov EL, Shirinsky VP. Western Blotting-Based Quantitative Measurement of Myosin II Regulatory Light Chain Phosphorylation in Small Amounts of Non-muscle Cells. BIOCHEMISTRY (MOSCOW) 2019; 84:11-19. [DOI: 10.1134/s0006297919010024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
Lin J, He Y, Chen L, Chen X, Zang S, Lin W. MYLK promotes hepatocellular carcinoma progression through regulating cytoskeleton to enhance epithelial-mesenchymal transition. Clin Exp Med 2018; 18:523-533. [PMID: 29855744 DOI: 10.1007/s10238-018-0509-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/24/2018] [Indexed: 11/24/2022]
Abstract
Myosin light chain kinase (MYLK) is found to catalyze the phosphorylation of myosin light chains (MLC) and regulate invasion and metastasis in some malignancies. However, there is little knowledge on the role of MYLK in hepatocellular carcinoma (HCC), and no studies have been conducted to investigate the mechanisms underlying MYLK-mediated promotion of HCC invasion and metastasis until now. In this study, we investigated the expression of MYLK in 50 pairs of human HCC and adjacent liver specimens. High MYLK expression was significantly correlated with aggressive clinicopathological features including tumor encapsulation, microvascular invasion and metastasis. In vitro assays showed that shRNA-induced MYLK knockdown significantly inhibited the wound-healing ability of HCC cells and the ability to migrate and invade through Matrigel. We next uncovered that MYLK knockdown resulted in a reduction in the number of F-actin stress fibers, disorganization of F-actin architectures and morphological alterations of HCC cells. Phosphorylated MLC, rather than total MLC, was found to be markedly reduced in response to downregulation of MYLK expression, and MYLK-regulated actin cytoskeleton through phosphorylating MLC in HCC cells. In addition, Western blotting assay revealed downregulation of the epithelial marker E-cadherin and upregulation of mesenchymal markers Vimentin, N-cadherin and Snail. Taken together, our findings indicate that MYLK promotes HCC progression by altering cytoskeleton to enhance epithelial-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Jie Lin
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, 350001, China.,Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Yihui He
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, 350001, China.,Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Lingfeng Chen
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, 350001, China.,Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Xiaoyan Chen
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, 350001, China.,Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Shengbing Zang
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Wansong Lin
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, No.420, Fuma Road, Jinan District, Fuzhou City, 350014, Fujian Province, China. .,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China.
| |
Collapse
|
26
|
Zhu L, Li C, Du G, Pan M, Liu G, Pan W, Li X. High glucose upregulates myosin light chain kinase to induce microfilament cytoskeleton rearrangement in hippocampal neurons. Mol Med Rep 2018; 18:216-222. [PMID: 29749555 PMCID: PMC6059672 DOI: 10.3892/mmr.2018.8960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic hyperglycemia leads to myosin light chain kinase (MLCK) upregulation and induces neuronal damage. However, the underlying molecular mechanism of neuronal damage in hyperglycemia has not yet been fully elucidated. In the present study, hippocampal neuronal cells were cultured and treated with a high glucose concentration (45 mmol/l). The results demonstrated that high glucose induced shrinking of the synapses, nuclear shape irregularity and microfilament damage. Filamentous actin (F‑actin) filaments were rearranged, cell apoptosis rate was increased and the protein expression of MLCK and phosphorylated (p)‑MLC was upregulated. The MLCK inhibitor ML‑7 largely reversed the alterations in the microfilament cytoskeleton, inhibited F‑actin depolymerization, reduced apoptosis and downregulated MLCK and p‑MLC protein expression. Overall, these results indicated that high glucose upregulated MLCK to promote F‑actin depolymerization, which induced microfilament cytoskeleton rearrangement in hippocampal neuronal cells.
Collapse
Affiliation(s)
- Liying Zhu
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Chengcheng Li
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Guiqin Du
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Meixiu Pan
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Guoqi Liu
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Wei Pan
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Xing Li
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
27
|
Huang C, Zhang Z, Wang L, Liu J, Gong X, Zhang C. ML-7 attenuates airway inflammation and remodeling via inhibiting the secretion of Th2 cytokines in mice model of asthma. Mol Med Rep 2018; 17:6293-6300. [PMID: 29512725 PMCID: PMC5928606 DOI: 10.3892/mmr.2018.8683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/01/2017] [Indexed: 01/05/2023] Open
Abstract
Previous studies have indicated that smooth muscle myosin light chain kinase (MLCK) has a prominent role in the regulation of smooth muscle contraction, which tends to be upregulated in asthma. In recent years, numerous studies have reported that MLCK is intimately connected with the immunoregulatory mechanism of T cells. The imbalance of T helper type 1 cells (Th1)/Th2 constitutes the immune-associated pathological basis of chronic asthma. Th2-associated cytokines, including interleukin-4, −5, −13, −25 and −33, are involved in airway inflammation, hyperresponsiveness and remodeling, which leads to a progressive decline in lung function. The purpose of the present study was to verify whether inhibition of bronchial MLCK attenuated the expression Th2-associated cytokines in asthmatic mice, including the above-mentioned ones. Female BALB/c mice were used to establish an ovalbumin (OVA)-induced model of asthma, of which one group was treated with the MLCK inhibitor (5-iodonaphthalene-1-sulfonyl) homopiperazine (ML-7). The inhibitor of MLCK, ML-7 attenuated airway inflammation and remodeling by reducing inflammatory cell infiltration and the secretion of Th2 cytokines in mice model of asthma, which may represent a promising therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Chuanjun Huang
- Department of Respiratory Diseases, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, P.R. China
| | - Zewen Zhang
- Department of Medical Imaging and Nuclear Medicine, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Liuxin Wang
- Department of Respiratory Medicine, The First People's Hospital of Jining, Jining, Shandong 272011, P.R. China
| | - Ju Liu
- Department of Medical Research Center, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Xiaodan Gong
- Department of Respiratory Diseases, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Caiqing Zhang
- Department of Respiratory Diseases, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|