1
|
Lateef OM, Foote C, Power G, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. LIM kinases in cardiovascular health and disease. Front Physiol 2024; 15:1506356. [PMID: 39744707 PMCID: PMC11688343 DOI: 10.3389/fphys.2024.1506356] [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: 10/05/2024] [Accepted: 11/28/2024] [Indexed: 01/14/2025] Open
Abstract
The Lim Kinase (LIMK) family of serine/threonine kinases is comprised of LIMK1 and LIMK2, which are central regulators of cytoskeletal dynamics via their well-characterized roles in promoting actin polymerization and destabilizing the cellular microtubular network. The LIMKs have been demonstrated to modulate several fundamental physiological processes, including cell cycle progression, cell motility and migration, and cell differentiation. These processes play important roles in maintaining cardiovascular health. However, LIMK activity in healthy and pathological states of the cardiovascular system is poorly characterized. This review highlights the cellular and molecular mechanisms involved in LIMK activation and inactivation, examining its roles in the pathophysiology of vascular and cardiac diseases such as hypertension, aneurysm, atrial fibrillation, and valvular heart disease. It addresses the LIMKs' involvement in processes that support cardiovascular health, including vasculogenesis, angiogenesis, and endothelial mechanotransduction. The review also features how LIMK activity participates in endothelial cell, vascular smooth muscle cell, and cardiomyocyte physiology and its implications in pathological states. A few recent preclinical studies demonstrate the therapeutic potential of LIMK inhibition. We conclude by proposing that future research should focus on the potential clinical relevance of LIMK inhibitors as therapeutic agents to reduce the burden of cardiovascular disease and improve patient outcomes.
Collapse
Affiliation(s)
- Olubodun M. Lateef
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, University of Missouri Columbia, Columbia, MO, United States
| | - Christopher Foote
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Gavin Power
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Camila Manrique-Acevedo
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Columbia, MO, United States
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
| | - Luis A. Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, University of Missouri Columbia, Columbia, MO, United States
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
2
|
Sheta YS, Sarg MT, Abdulrahman FG, Nossier ES, Husseiny EM. Novel imidazolone derivatives as potential dual inhibitors of checkpoint kinases 1 and 2: Design, synthesis, cytotoxicity evaluation, and mechanistic insights. Bioorg Chem 2024; 149:107471. [PMID: 38823311 DOI: 10.1016/j.bioorg.2024.107471] [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: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Applying various drug design strategies including ring variation, substituents variation, and ring fusion, two series of 2-(alkylthio)-5-(arylidene/heteroarylidene)imidazolones and imidazo[1,2-a]thieno[2,3-d]pyrimidines were designed and prepared as dual potential Chk1 and Chk2 inhibitors. The newly synthesized hybrids were screened in NCI 60 cell line panel where the most active derivatives 4b, d-f, and 6a were further estimated for their five dose antiproliferative activity against the most sensitive tumor cells including breast MCF-7 and MDA-MB-468 and non-small cell lung cancer EKVX as well as normal WI-38 cell. Noticeably, increasing the carbon chain attached to thiol moiety at C-2 of imidazolone scaffold elevated the cytotoxic activity. Hence, compounds 4e and 4f, containing S-butyl fragment, exhibited the most antiproliferative activity against the tested cells where 4f showed extremely potent selectivity toward them. As well, compound 6a, containing imidazothienopyrimidine core, exerted significant cytotoxic activity and selectivity toward the examined cells. The mechanistic investigation of the most active cytotoxic analogs was achieved through the evaluation of their inhibitory activity against Chk1 and Chk2. Results revealed that 4f displayed potent dual inhibition of both Chk1 and Chk2 with IC50 equal 0.137 and 0.25 μM, respectively. It also promoted its antiproliferative and Chk suppression activity via EKVX cell cycle arrest at S phase through stimulating the apoptotic approach. The apoptosis induction was also emphasized by elevating the expression of Caspase-3 and Bax, that are accompanied by Bcl-2 diminution. The in silico molecular docking and ADMET profiles of the most active analogs have been carried out to evaluate their potential as significant anticancer drug candidates.
Collapse
Affiliation(s)
- Yasmin S Sheta
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Marwa T Sarg
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Fatma G Abdulrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Eman S Nossier
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt; The National Committee of Drugs, Academy of Scientific Research and Technology, Cairo 11516, Egypt
| | - Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt.
| |
Collapse
|
3
|
Chong ZX, Ho WY, Yeap SK. Decoding the tumour-modulatory roles of LIMK2. Life Sci 2024; 347:122609. [PMID: 38580197 DOI: 10.1016/j.lfs.2024.122609] [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: 12/13/2023] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
LIM domains kinase 2 (LIMK2) is a 72 kDa protein that regulates actin and cytoskeleton reorganization. Once phosphorylated by its upstream activator (ROCK1), LIMK2 can phosphorylate cofilin to inactivate it. This relieves the levering stress on actin and allows polymerization to occur. Actin rearrangement is essential in regulating cell cycle progression, apoptosis, and migration. Dysregulation of the ROCK1/LIMK2/cofilin pathway has been reported to link to the development of various solid cancers such as breast, lung, and prostate cancer and liquid cancer like leukemia. This review aims to assess the findings from multiple reported in vitro, in vivo, and clinical studies on the potential tumour-regulatory role of LIMK2 in different human cancers. The findings of the selected literature unraveled that activated AKT, EGF, and TGF-β pathways can upregulate the activities of the ROCK1/LIMK2/cofilin pathway. Besides cofilin, LIMK2 can modulate the cellular levels of other proteins, such as TPPP1, to promote microtubule polymerization. The tumour suppressor protein p53 can transactivate LIMK2b, a splice variant of LIMK2, to induce cell cycle arrest and allow DNA repair to occur before the cell enters the next phase of the cell cycle. Additionally, several non-coding RNAs, such as miR-135a and miR-939-5p, could also epigenetically regulate the expression of LIMK2. Since the expression of LIMK2 is dysregulated in several human cancers, measuring the tissue expression of LIMK2 could potentially help diagnose cancer and predict patient prognosis. As LIMK2 could play tumour-promoting and tumour-inhibiting roles in cancer development, more investigation should be conducted to carefully evaluate whether introducing a LIMK2 inhibitor in cancer patients could slow cancer progression without posing clinical harms.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia.
| |
Collapse
|
4
|
Zhang L, Zhang X, Shi Y, Ni Y, Fei J, Jin Z, Li W, Wang X, Wu N. Role and potential therapeutic value of histone methyltransferases in drug resistance mechanisms in lung cancer. Front Oncol 2024; 14:1376916. [PMID: 38525426 PMCID: PMC10957659 DOI: 10.3389/fonc.2024.1376916] [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: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/26/2024] Open
Abstract
Lung cancer, ranking second globally in both incidence and high mortality among common malignant tumors, presents a significant challenge with frequent occurrences of drug resistance despite the continuous emergence of novel therapeutic agents. This exacerbates disease progression, tumor recurrence, and ultimately leads to poor prognosis. Beyond acquired resistance due to genetic mutations, mounting evidence suggests a critical role of epigenetic mechanisms in this process. Numerous studies have indicated abnormal expression of Histone Methyltransferases (HMTs) in lung cancer, with the abnormal activation of certain HMTs closely linked to drug resistance. HMTs mediate drug tolerance in lung cancer through pathways involving alterations in cellular metabolism, upregulation of cancer stem cell-related genes, promotion of epithelial-mesenchymal transition, and enhanced migratory capabilities. The use of HMT inhibitors also opens new avenues for lung cancer treatment, and targeting HMTs may contribute to reversing drug resistance. This comprehensive review delves into the pivotal roles and molecular mechanisms of HMTs in drug resistance in lung cancer, offering a fresh perspective on therapeutic strategies. By thoroughly examining treatment approaches, it provides new insights into understanding drug resistance in lung cancer, supporting personalized treatment, fostering drug development, and propelling lung cancer therapy into novel territories.
Collapse
Affiliation(s)
- Linxiang Zhang
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xueying Zhang
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yan Shi
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuhan Ni
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jiaojiao Fei
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhixin Jin
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Wenjuan Li
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaojing Wang
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Nan Wu
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
5
|
Wollscheid HP, Ulrich HD. Chromatin meets the cytoskeleton: the importance of nuclear actin dynamics and associated motors for genome stability. DNA Repair (Amst) 2023; 131:103571. [PMID: 37738698 DOI: 10.1016/j.dnarep.2023.103571] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
The actin cytoskeleton is of fundamental importance for numerous cellular processes, including intracellular transport, cell plasticity, and cell migration. However, functions of filamentous actin (F-actin) in the nucleus remain understudied due to the comparatively low abundance of nuclear actin and the resulting experimental limitations to its visualization. Owing to recent technological advances such as super-resolution microscopy and the development of nuclear-specific actin probes, essential roles of the actin cytoskeleton in the context of genome maintenance are now emerging. In addition to the contributions of monomeric actin as a component of multiple important nuclear protein complexes, nuclear actin has been found to undergo polymerization in response to DNA damage and DNA replication stress. Consequently, nuclear F-actin plays important roles in the regulation of intra-nuclear mobility of repair and replication foci as well as the maintenance of nuclear shape, two important aspects of efficient stress tolerance. Beyond actin itself, there is accumulating evidence for the participation of multiple actin-binding proteins (ABPs) in the surveillance of genome integrity, including nucleation factors and motor proteins of the myosin family. Here we summarize recent findings highlighting the importance of actin cytoskeletal factors within the nucleus in key genome maintenance pathways.
Collapse
Affiliation(s)
- Hans-Peter Wollscheid
- Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, Mainz D - 55128, Germany.
| | - Helle D Ulrich
- Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, Mainz D - 55128, Germany.
| |
Collapse
|
6
|
Jiang X, Xu Z, Jiang S, Wang H, Xiao M, Shi Y, Wang K. PDZ and LIM Domain-Encoding Genes: Their Role in Cancer Development. Cancers (Basel) 2023; 15:5042. [PMID: 37894409 PMCID: PMC10605254 DOI: 10.3390/cancers15205042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
PDZ-LIM family proteins (PDLIMs) are a kind of scaffolding proteins that contain PDZ and LIM interaction domains. As protein-protein interacting molecules, PDZ and LIM domains function as scaffolds to bind to a variety of proteins. The PDLIMs are composed of evolutionarily conserved proteins found throughout different species. They can participate in cell signal transduction by mediating the interaction of signal molecules. They are involved in many important physiological processes, such as cell differentiation, proliferation, migration, and the maintenance of cellular structural integrity. Studies have shown that dysregulation of the PDLIMs leads to tumor formation and development. In this paper, we review and integrate the current knowledge on PDLIMs. The structure and function of the PDZ and LIM structural domains and the role of the PDLIMs in tumor development are described.
Collapse
Affiliation(s)
| | | | | | | | | | - Yueli Shi
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; (X.J.); (Z.X.); (S.J.); (H.W.); (M.X.)
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; (X.J.); (Z.X.); (S.J.); (H.W.); (M.X.)
| |
Collapse
|
7
|
Shah K, Cook M. LIMK2: A Multifaceted kinase with pleiotropic roles in human physiology and pathologies. Cancer Lett 2023; 565:216207. [PMID: 37141984 PMCID: PMC10316521 DOI: 10.1016/j.canlet.2023.216207] [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: 03/23/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023]
Abstract
LIMK2, a serine-specific kinase, was discovered as an actin dynamics regulating kinase. Emerging studies have shown its pivotal role in numerous human malignancies and neurodevelopmental disorder. Inducible knockdown of LIMK2 fully reverses tumorigenesis, underscoring its potential as a clinical target. However, the molecular mechanisms leading to its upregulation and its deregulated activity in various diseases largely remain unknown. Similarly, LIMK2's peptide substrate specificity has not been analyzed. This is particularly important for LIMK2, a kinase almost three decades old, as only a handful of its substrates are known to date. As a result, most of LIMK2's physiological and pathological roles have been assigned to its regulation of actin dynamics via cofilin. This review focuses on LIMK2's unique catalytic mechanism, substrate specificity and its upstream regulators at transcriptional, post-transcriptional and post-translational stages. Moreover, emerging studies have unveiled a few tumor suppressors and oncogenes as LIMK2's direct substrates, which in turn have uncovered novel molecular mechanisms by which it plays pleiotropic roles in human physiology and pathologies independent of actin dynamics.
Collapse
Affiliation(s)
- Kavita Shah
- Department of Chemistry and Purdue University Center for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| | - Mason Cook
- Department of Chemistry and Purdue University Center for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA
| |
Collapse
|
8
|
Magalhaes YT, Boell VK, Cardella GD, Forti FL. Downregulation of the Rho GTPase pathway abrogates resistance to ionizing radiation in wild-type p53 glioblastoma by suppressing DNA repair mechanisms. Cell Death Dis 2023; 14:283. [PMID: 37085490 PMCID: PMC10121706 DOI: 10.1038/s41419-023-05812-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/09/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023]
Abstract
Glioblastoma (GBM), the most common aggressive brain tumor, is characterized by rapid cellular infiltration and is routinely treated with ionizing radiation (IR), but therapeutic resistance inevitably recurs. The actin cytoskeleton of glioblastoma cells provides their high invasiveness, but it remains unclear whether Rho GTPases modulate DNA damage repair and therapeutic sensitivity. Here, we irradiated glioblastoma cells with different p53 status and explored the effects of Rho pathway inhibition to elucidate how actin cytoskeleton disruption affects the DNA damage response and repair pathways. p53-wild-type and p53-mutant cells were subjected to Rho GTPase pathway modulation by treatment with C3 toxin; knockdown of mDia-1, PFN1 and MYPT1; or treatment with F-actin polymerization inhibitors. Rho inhibition increased the sensitivity of glioma cells to IR by increasing the number of DNA double-strand breaks and delaying DNA repair by nonhomologous end-joining in p53-wild-type cells. p53 knockdown reversed this phenotype by reducing p21 expression and Rho signaling activity, whereas reactivation of p53 in p53-mutant cells by treatment with PRIMA-1 reversed these effects. The interdependence between p53 and Rho is based on nuclear p53 translocation facilitated by G-actin and enhanced by IR. Isolated IR-resistant p53-wild-type cells showed an altered morphology and increased stress fiber formation: inhibition of Rho or actin polymerization decreased cell viability in a p53-dependent manner and reversed the resistance phenotype. p53 silencing reversed the Rho inhibition-induced sensitization of IR-resistant cells. Rho inhibition also impaired the repair of IR-damaged DNA in 3D spheroid models. Rho GTPase activity and actin cytoskeleton dynamics are sensitive targets for the reversal of acquired resistance in GBM tumors with wild-type p53.
Collapse
Affiliation(s)
- Yuli Thamires Magalhaes
- Laboratory of Biomolecular Systems Signaling, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Viktor Kalbermatter Boell
- Laboratory of Biomolecular Systems Signaling, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Giovanna Duo Cardella
- Laboratory of Biomolecular Systems Signaling, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Fabio Luis Forti
- Laboratory of Biomolecular Systems Signaling, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
9
|
Villalonga E, Mosrin C, Normand T, Girardin C, Serrano A, Žunar B, Doudeau M, Godin F, Bénédetti H, Vallée B. LIM Kinases, LIMK1 and LIMK2, Are Crucial Node Actors of the Cell Fate: Molecular to Pathological Features. Cells 2023; 12:cells12050805. [PMID: 36899941 PMCID: PMC10000741 DOI: 10.3390/cells12050805] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
LIM kinase 1 (LIMK1) and LIM kinase 2 (LIMK2) are serine/threonine and tyrosine kinases and the only two members of the LIM kinase family. They play a crucial role in the regulation of cytoskeleton dynamics by controlling actin filaments and microtubule turnover, especially through the phosphorylation of cofilin, an actin depolymerising factor. Thus, they are involved in many biological processes, such as cell cycle, cell migration, and neuronal differentiation. Consequently, they are also part of numerous pathological mechanisms, especially in cancer, where their involvement has been reported for a few years and has led to the development of a wide range of inhibitors. LIMK1 and LIMK2 are known to be part of the Rho family GTPase signal transduction pathways, but many more partners have been discovered over the decades, and both LIMKs are suspected to be part of an extended and various range of regulation pathways. In this review, we propose to consider the different molecular mechanisms involving LIM kinases and their associated signalling pathways, and to offer a better understanding of their variety of actions within the physiology and physiopathology of the cell.
Collapse
Affiliation(s)
- Elodie Villalonga
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Christine Mosrin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Thierry Normand
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Caroline Girardin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Amandine Serrano
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Bojan Žunar
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Michel Doudeau
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Fabienne Godin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Hélène Bénédetti
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Béatrice Vallée
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
- Correspondence: ; Tel.: +33-(0)2-38-25-76-11
| |
Collapse
|
10
|
Su Y, Xu B, Shen Q, Lei Z, Zhang W, Hu T. LIMK2 Is a Novel Prognostic Biomarker and Correlates With Tumor Immune Cell Infiltration in Lung Squamous Cell Carcinoma. Front Immunol 2022; 13:788375. [PMID: 35273591 PMCID: PMC8902256 DOI: 10.3389/fimmu.2022.788375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Previous research found that LIM domain kinase 2 (LIMK2) expression correlated with a poor prognosis in many cancers. However, its role in lung squamous cell carcinoma (LUSC) has not yet been clarified. Our study aimed to clarify the role of LIMK2 in LUSC prognosis prediction and explore the relationship between LIMK2 and immune infiltration in LUSC. In this study, we first analyzed the expression level and prognostic value of LIMK2 across cancers. Subsequently, we explored the association of LIMK2 expression with immune infiltrating cells and immune checkpoints. our study found that LIMK2 was highly expressed and positively associated with the overall survival of LUSC. Moreover, our study further indicated that LIMK2 expression was significantly negatively correlated with immune cell infiltration and immune checkpoints in LUSC. Finally, we confirmed upstream regulatory noncoding RNAs (ncRNAs) of LIMK2, and the PVT1 and DHRS4-AS1/miR-423-5p/LIMK2 regulatory axes were successfully constructed in LUSC. Put together, LIMK2 is a novel prognostic biomarker and correlates with tumor immune cell infiltration in LUSC, and the expression of LIMK2 is regulated by the PVT1 and DHRS4-AS1/miR-423-5p axes.
Collapse
Affiliation(s)
- Yongcheng Su
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China
| | - Beibei Xu
- Department of General Surgery, The First Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qianwen Shen
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China
| | - Ziyu Lei
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China
| | - Wenqing Zhang
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China
| | - Tianhui Hu
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China.,Shenzhen Research Institute of Xiamen University, Shenzhen, China
| |
Collapse
|
11
|
Magalhaes YT, Farias JO, Silva LE, Forti FL. GTPases, genome, actin: A hidden story in DNA damage response and repair mechanisms. DNA Repair (Amst) 2021; 100:103070. [PMID: 33618126 DOI: 10.1016/j.dnarep.2021.103070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022]
Abstract
The classical small Rho GTPase (Rho, Rac, and Cdc42) protein family is mainly responsible for regulating cell motility and polarity, membrane trafficking, cell cycle control, and gene transcription. Cumulative recent evidence supports important roles for these proteins in the maintenance of genomic stability. Indeed, DNA damage response (DDR) and repair mechanisms are some of the prime biological processes that underlie several disease phenotypes, including genetic disorders, cancer, senescence, and premature aging. Many reports guided by different experimental approaches and molecular hypotheses have demonstrated that, to some extent, direct modulation of Rho GTPase activity, their downstream effectors, or actin cytoskeleton regulation contribute to these cellular events. Although much attention has been paid to this family in the context of canonical actin cytoskeleton remodeling, here we provide a contextualized review of the interplay between Rho GTPase signaling pathways and the DDR and DNA repair signaling components. Interesting questions yet to be addressed relate to the spatiotemporal dynamics of this collective response and whether it correlates with different subcellular pools of Rho GTPases. We highlight the direct and indirect targets, some of which still lack experimental validation data, likely associated with Rho GTPase activation that provides compelling evidence for further investigation in DNA damage-associated events and with potential therapeutic applications in translational medicine.
Collapse
Affiliation(s)
- Yuli T Magalhaes
- Laboratory of Biomolecular Systems Signaling, Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, Brazil
| | - Jessica O Farias
- Laboratory of Biomolecular Systems Signaling, Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, Brazil
| | - Luiz E Silva
- Laboratory of Biomolecular Systems Signaling, Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, Brazil
| | - Fabio L Forti
- Laboratory of Biomolecular Systems Signaling, Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, Brazil.
| |
Collapse
|
12
|
Ma J, Liu X, Liu P, Lu W, Shen X, Ma R, Zong H. Identification of a new p53 responsive element in the promoter region of anillin. Int J Mol Med 2020; 45:1563-1570. [PMID: 32323752 DOI: 10.3892/ijmm.2020.4527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/06/2019] [Indexed: 11/06/2022] Open
Abstract
The expression of anillin mRNA and protein is regulated in a cell cycle‑dependent manner. However, the mechanism underlying this process is unclear. Previous studies analyzing the sequence of the 5'‑untranslated region of anillin have unveiled several putative p53 binding sites. Therefore, the present study hypothesized that the anillin gene may be repressed by p53 and that the commonly observed mutation (or loss of function) of p53 may serve a role in this phenotype. Bioinformatic analysis of the anillin promoter region revealed potential p53 responsive elements. Of those identified, 2 were able to bind p53 protein, as determined via a chromatin immunoprecipitation assay. Although it was hypothesized that DNA damage and resultant p53 expression would repress anillin expression, the results revealed that anillin mRNA and protein expression levels were negatively regulated by DNA damage in the wild‑type p53 cells, but not in the isogenic p53 null cells. Furthermore, DNA sequences encompassing the p53 binding site downregulated luciferase transgenes in a p53 dependent manner. Taken together, these data indicated that anillin was negatively regulated by p53 and that anillin overexpression observed in cancer may be a p53‑mediated phenomenon. The data from the present study provided further evidence for the role of p53 in the biologically crucial process of cytokinesis.
Collapse
Affiliation(s)
- Jiao Ma
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiaotong University Medical School, Shanghai 200025, P.R. China
| | - Xinying Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiaotong University Medical School, Shanghai 200025, P.R. China
| | - Pengyi Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiaotong University Medical School, Shanghai 200025, P.R. China
| | - Wenqing Lu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiaotong University Medical School, Shanghai 200025, P.R. China
| | - Xinxin Shen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiaotong University Medical School, Shanghai 200025, P.R. China
| | - Ruixiang Ma
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiaotong University Medical School, Shanghai 200025, P.R. China
| | - Hongliang Zong
- Shanghai PerHum Therapeutics Co. Ltd., Shanghai 200052, P.R. China
| |
Collapse
|
13
|
Sirt3 Exerts Its Tumor-Suppressive Role by Increasing p53 and Attenuating Response to Estrogen in MCF-7 Cells. Antioxidants (Basel) 2020; 9:antiox9040294. [PMID: 32244715 PMCID: PMC7222218 DOI: 10.3390/antiox9040294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/21/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Estrogen (E2) is a major risk factor for the initiation and progression of malignancy in estrogen receptor (ER) positive breast cancers, whereas sirtuin 3 (Sirt3), a major mitochondrial NAD+-dependent deacetylase, has the inhibitory effect on the tumorigenic properties of ER positive MCF-7 breast cancer cells. Since it is unclear if this effect is mediated through the estrogen receptor alpha (ERα) signaling pathway, in this study, we aimed to determine if the tumor-suppressive function of Sirt3 in MCF-7 cells interferes with their response to E2. Although we found that Sirt3 improves the antioxidative response and mitochondrial fitness of the MCF-7 cells, it also increases DNA damage along with p53, AIF, and ERα expression. Moreover, Sirt3 desensitizes cells to the proliferative effect of E2, affects p53 by disruption of the ERα–p53 interaction, and decreases proliferation, colony formation, and migration of the cells. Our observations indicate that these tumor-suppressive effects of Sirt3 could be reversed by E2 treatment only to a limited extent which is not sufficient to recover the tumorigenic properties of the MCF-7 cells. This study provides new and interesting insights with respect to the functional role of Sirt3 in the E2-dependent breast cancers.
Collapse
|
14
|
Li B, Zhou P, Xu K, Chen T, Jiao J, Wei H, Yang X, Xu W, Wan W, Xiao J. Metformin induces cell cycle arrest, apoptosis and autophagy through ROS/JNK signaling pathway in human osteosarcoma. Int J Biol Sci 2020; 16:74-84. [PMID: 31892847 PMCID: PMC6930379 DOI: 10.7150/ijbs.33787] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 10/13/2019] [Indexed: 01/07/2023] Open
Abstract
Metformin, an ancient drug commonly used for treating type II diabetes, has been associated to anti-cancer capacity in a variety of developing cancers, though the mechanism remains elusive. Here, we aimed to examine the inhibitory effect of metformin in osteosarcoma. Herein, we demonstrated that metformin treatment blocked proliferation progression by causing accumulation of G2/M phase in U2OS and 143B cells. Furthermore, metformin treatment triggered programmed cell death process in osteosarcoma cell lines. Further research indicated the induction of apoptosis and autophagy triggered by metformin could remarkably attenuate after the treatment of ROS scavenger NAC and JNK inhibitor SP600125. Additionally, our results showed that NAC-suppressed JNK/c-Jun signaling pathway could have been activated through metformin treatment. Lastly, metformin could inhibit osteosarcoma growth under safe dose in vivo. Thus, we propose that metformin could induce cell cycle arrest as well as programmed cell death, including apoptosis and autophagy, through ROS-dependent JNK/c-Jun cascade in human osteosarcoma. This metformin-induced pathway provides further insights into its antitumor potential molecular mechanism and illuminates potential cancer targets for osteosarcoma.
Collapse
Affiliation(s)
- Bo Li
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Pingting Zhou
- Department of Radiation Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kehan Xu
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tianrui Chen
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jian Jiao
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Haifeng Wei
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xinghai Yang
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wei Xu
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wei Wan
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jianru Xiao
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
15
|
Xu M, Wang F, Li G, Wang X, Fang X, Jin H, Chen Z, Zhang J, Fu L. MED12 exerts an emerging role in actin-mediated cytokinesis via LIMK2/cofilin pathway in NSCLC. Mol Cancer 2019; 18:93. [PMID: 31072327 PMCID: PMC6509838 DOI: 10.1186/s12943-019-1020-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/22/2019] [Indexed: 12/31/2022] Open
Abstract
Background Mediator complex subunit 12 (MED12) is an essential hub for transcriptional regulation, in which mutations and overexpression were reported to be associated with several kinds of malignancies. Nevertheless, the role of MED12 in non-small cell lung cancer (NSCLC) remains to be elucidated. Methods MED12 mutation was detected by Next-generation sequencing. The expression of MED12 in 179 human NSCLC tissue samples and 73 corresponding adjacent normal lung tissue samples was measured by immunohistochemistry (IHC). CRISPR-Cas9 was used to knock out MED12 in PC9 and SPC-A1 cells. MED12 rescued stable cell lines were generated by lentivirus infection. We traced cell division process by live cell imaging. The molecular mechanism of aborted cytokinesis resulted by MED12 knockout was investigated by RNA-seq. Effects of MED12 deletion on the proliferation of NSCLC cells were determined by MTT assay and Colony-formation assay in vitro and xenograft tumor model in nude mouse. Cell senescence was measured by SA-β-gal staining. Results In our study, no MED12 exon mutation was detected in NSCLC samples, whereas we found that MED12 was overexpressed in human NSCLC tissues, which positively correlated with the tumor volume and adversely affected patient survival. Furthermore, knockout MED12 in NSCLC cell lines resulted in cytokinesis failure, displayed a multinuclear phenotype, and disposed to senescence, and become non-viable. Lack of MED12 decreased the proliferative potential of NSCLC cells and limited the tumor growth in vivo. Mechanism investigations revealed that MED12 knockout activated LIMK2, caused aberrant actin cytoskeleton remodeling, and disrupted the abscission of intercellular bridge, which led to the cytokinesis failure. Reconstitution of exogenous MED12 restored actin dynamics, normal cytokinesis and cell proliferation capacity in MED12 knockout cells. Conclusions These results revealed a novel role of MED12 as an important regulator for maintaining accurate cytokinesis and survival in NSCLC cells, which may offer a therapeutic strategy to control tumor growth for NSCLC patients especially those highly expressed MED12. Electronic supplementary material The online version of this article (10.1186/s12943-019-1020-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meng Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No.651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China.,Radiotherapy Department of Thorax & Abdomen Tumor, Cancer Center, The First People's Hospital of Foshan Affiliated to Sun Yat-sen University, Foshan, 528000, China
| | - Fang Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No.651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China
| | - Guibo Li
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, 518083, China
| | - Xiaokun Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No.651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China
| | - Xiaona Fang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No.651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China
| | - Haoxuan Jin
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, 518083, China
| | - Zhen Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No.651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China
| | - Jianye Zhang
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Liwu Fu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No.651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China.
| |
Collapse
|
16
|
LIMK2-1, a new isoform of human LIMK2, regulates actin cytoskeleton remodeling via a different signaling pathway than that of its two homologs, LIMK2a and LIMK2b. Biochem J 2018; 475:3745-3761. [DOI: 10.1042/bcj20170961] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 12/29/2022]
Abstract
LIMK1 and LIMK2 (LIMKs, LIM kinases) are kinases that play a crucial role in cytoskeleton dynamics by independently regulating both actin filament and microtubule remodeling. LIMK1 and, more recently, LIMK2 have been shown to be involved in cancer development and metastasis, resistance of cancer cells to microtubule-targeted treatments, neurological diseases, and viral infection. LIMKs have thus recently emerged as new therapeutic targets. Databanks describe three isoforms of human LIMK2: LIMK2a, LIMK2b, and LIMK2-1. Evidence suggests that they may not have completely overlapping functions. We biochemically characterized the three isoforms to better delineate their potential roles, focusing on LIMK2-1, which has only been described at the mRNA level in a single study. LIMK2-1 has a protein phosphatase 1 (PP1) inhibitory domain at its C-terminus which its two counterparts do not. We showed that the LIMK2-1 protein is indeed synthesized. LIMK2-1 does not phosphorylate cofilin, the canonical substrate of LIMKs, although it has kinase activity and promotes actin stress fiber formation. Instead, it interacts with PP1 and partially inhibits its activity towards cofilin. Our data suggest that LIMK2-1 regulates actin cytoskeleton dynamics by preventing PP1-mediated cofilin dephosphorylation, rather than by directly phosphorylating cofilin as its two counterparts, LIMK2a and LIMK2b. This specificity may allow for tight regulation of the phospho-cofilin pool, determining the fate of the cell.
Collapse
|
17
|
Wang W, Yang C, Nie H, Qiu X, Zhang L, Xiao Y, Zhou W, Zeng Q, Zhang X, Wu Y, Liu J, Ying M. LIMK2 acts as an oncogene in bladder cancer and its functional SNP in the microRNA-135a binding site affects bladder cancer risk. Int J Cancer 2018; 144:1345-1355. [PMID: 30006972 PMCID: PMC6587996 DOI: 10.1002/ijc.31757] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 04/16/2018] [Accepted: 06/14/2018] [Indexed: 01/02/2023]
Abstract
LIM kinases modulate multiple aspects of cancer development, including cell proliferation and survival. As the mechanisms of LIMK-associated tumorigenesis are still unclear, we analyzed the tumorigenic functions of LIM kinase 2 (LIMK2) in human bladder cancer (BC) and explored whether the newly identified LIMK2 3´-UTR SNP rs2073859 (G-to-A allele) is correlated with clinical features. Expression levels of LIMK2 in 38 human BC tissues and eight cell lines were examined using quantitative real-time PCR and immunohistochemistry. LIMK2 was overexpressed in most BC tissues (27/38, 71%) and BC-derived cell lines (6/8), and was more frequently overexpessed in high-grade than low-grade BC (80% vs. 47%). The effects of LIMK2 on BC cell proliferation, survival and migration, were studied by overexpression and RNA interference approaches in vitro and in vivo. LIMK2 overexpression promoted proliferation, migration and invasion of BC cells, while LIMK2 depletion inhibited cell invasion and viability and induced growth arrest in vitro and in vivo. PCR-Restriction Fragment Length Polymorphism (RFLP) was used to genotype LIMK2 SNP rs2073859 and multivariate logistic regression applied to assess the relationship between allele frequency and clinical features in 139 BC patients. Functional analyses localized SNP rs2073859 within the microRNA-135a seed-binding region and revealed significantly lower LIMK2 G allele expression. The frequency of A genotypes (AG + AA) was higher in the BC group than normal controls and correlated with risks of high-grade and high-stage BC. In conclusion, LIMK2 may function as an oncogene in human BC, while allele-specific regulation by microRNA-135a may influence disease risk.
Collapse
Affiliation(s)
- Wei Wang
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Chenglin Yang
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Haibo Nie
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Xiaofu Qiu
- Department of Urology, Guangdong NO.2 Provincial People's Hospital, Guangzhou, China
| | - Lianbo Zhang
- Department of Plastic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuansong Xiao
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Wuer Zhou
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Qinsong Zeng
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Xiaoming Zhang
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Yigao Wu
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Jun Liu
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Min Ying
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| |
Collapse
|
18
|
Gao X, Mi Y, Guo N, Xu H, Jiang P, Zhang R, Xu L, Gou X. Glioma in Schizophrenia: Is the Risk Higher or Lower? Front Cell Neurosci 2018; 12:289. [PMID: 30233327 PMCID: PMC6129591 DOI: 10.3389/fncel.2018.00289] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022] Open
Abstract
Whether persons with schizophrenia have a higher or lower incidence of cancer has been discussed for a long time. Due to the complex mechanisms and characteristics of different types of cancer, it is difficult to evaluate the exact relationship between cancers and schizophrenia without considering the type of tumor. Schizophrenia, a disabling mental illness that is now recognized as a neurodevelopmental disorder, is more correlated with brain tumors, such as glioma, than other types of tumors. Thus, we mainly focused on the relationship between schizophrenia and glioma morbidity. Glioma tumorigenesis and schizophrenia may share similar mechanisms; gene/pathway disruption would affect neurodevelopment and reduce the risk of glioma. The molecular defects of disrupted-in-schizophrenia-1 (DISC1), P53, brain-derived neurotrophic factor (BDNF) and C-X-C chemokine receptors type 4 (CXCR4) involved in schizophrenia pathogenesis might play opposite roles in glioma development. Many microRNAs (miRNAs) such as miR-183, miR-9, miR-137 and miR-126 expression change may be involved in the cross talk between glioma prevalence and schizophrenia. Finally, antipsychotic drugs may have antitumor effects. All these factors show that persons with schizophrenia have a decreased incidence of glioma; therefore, epidemiological investigation and studies comparing genetic and epigenetic aberrations involved in both of these complex diseases should be performed. These studies can provide more insightful knowledge about glioma and schizophrenia pathophysiology and help to determine the target/strategies for the prevention and treatment of the two diseases.
Collapse
Affiliation(s)
- Xingchun Gao
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic Medical Sciences & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yajing Mi
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic Medical Sciences & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Na Guo
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic Medical Sciences & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Hao Xu
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic Medical Sciences & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Pengtao Jiang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic Medical Sciences & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Ruisan Zhang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic Medical Sciences & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Lixian Xu
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic Medical Sciences & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic Medical Sciences & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| |
Collapse
|
19
|
Galal SA, Khattab M, Shouman SA, Ramadan R, Kandil OM, Kandil OM, Tabll A, El Abd YS, El-Shenawy R, Attia YM, El-Rashedy AA, El Diwani HI. Part III: Novel checkpoint kinase 2 (Chk2) inhibitors; design, synthesis and biological evaluation of pyrimidine-benzimidazole conjugates. Eur J Med Chem 2018; 146:687-708. [PMID: 29407991 DOI: 10.1016/j.ejmech.2018.01.072] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 02/07/2023]
Abstract
Recently a dramatic development of the cancer drug discovery has been shown in the field of targeted cancer therapy. Checkpoint kinase 2 (Chk2) inhibitors offer a promising approach to enhance the effectiveness of cancer chemotherapy. Accordingly, in this study many pyrimidine-benzimidazole conjugates were designed and twelve feasible derivatives were selected to be synthesized to investigate their activity against Chk2 and subjected to study their antitumor activity alone and in combination with the genotoxic anticancer drugs cisplatin and doxorubicin on breast carcinoma, (ER+) cell line (MCF-7). The results indicated that the studied compounds inhibited Chk2 activity with high potency (IC50 = 5.56 nM - 46.20 nM). The studied candidates exhibited remarkable antitumor activity against MCF-7 (IG50 = 6.6 μM - 24.9 μM). Compounds 10a-c, 14 and 15 significantly potentiated the activity of the studied genotoxic drugs, whereas, compounds 9b and 20-23 antagonized their activity. Moreover, the combination of compound 10b with cisplatin revealed the best apoptotic effect as well as combination of compound 10b with doxorubicin led to complete arrest of the cell cycle at S phase where more than 40% of cells are in the S phase with no cells at G2/M. Structure-activity relationship was discussed on the basis of molecular modeling study using Molecular modeling Environment program (MOE).
Collapse
Affiliation(s)
- Shadia A Galal
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt.
| | - Muhammad Khattab
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt
| | - Samia A Shouman
- Department of Cancer Biology, National Cancer Institute, Cairo University, Egypt
| | - Raghda Ramadan
- Radiobiology Unit, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium; Department of Basic Medical Sciences, Physiology Group, Ghent University, Ghent, Belgium
| | - Omaima M Kandil
- Department of Animal Reproduction & Artificial Insemination, Division, of Veterinary Research, National Research Centre, Cairo, 12622, Egypt
| | - Omnia M Kandil
- Department of Parasitology, Animal Disease, Division, of Veterinary, National Research Centre, Cairo, 12622, Egypt
| | - Ashraf Tabll
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Yasmine S El Abd
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Reem El-Shenawy
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Yasmin M Attia
- Department of Cancer Biology, National Cancer Institute, Cairo University, Egypt
| | - Ahmed A El-Rashedy
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt
| | - Hoda I El Diwani
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt
| |
Collapse
|
20
|
Galal SA, Khairat SHM, Ali HI, Shouman SA, Attia YM, Ali MM, Mahmoud AE, Abdel-Halim AH, Fyiad AA, Tabll A, El-Shenawy R, El Abd YS, Ramdan R, El Diwani HI. Part II: New candidates of pyrazole-benzimidazole conjugates as checkpoint kinase 2 (Chk2) inhibitors. Eur J Med Chem 2018; 144:859-873. [PMID: 29316526 DOI: 10.1016/j.ejmech.2017.12.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/12/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023]
Abstract
The development of checkpoint kinase 2 (Chk2) inhibitors for the treatment of cancer has been an ongoing and attractive objective in drug discovery. In this study, twenty-one feasible pyrazole-benzimidazole conjugates were synthesized to study their effect against Chk2 activity using Checkpoint Kinase Assay. The antitumor activity of these compounds was investigated using SRB assay. A potentiation effect of the synthesized Chk2 inhibitors was also investigated using the genotoxic anticancer drugs cisplatin and doxorubicin on breast carcinoma, (ER+) cell line (MCF-7). In vivo Chk2 and antitumor activities of 8d as a single-agent, and in combination with doxorubicin, were evaluated in breast cancer bearing animals induced by N-methylnitrosourea. The effect of 8d alone and in combination with doxorubicin was also studied on cell-cycle phases of MCF-7 cells using flow cytometry analysis. The results revealed their potencies as Chk2 inhibitors with IC50 ranges from 9.95 to 65.07 nM. Generally the effect of cisplatin or doxorubicin was potentiated by the effect of most of the compounds that were studied. The in vivo results indicated that the combination of 8d and doxorubicin inhibited checkpoint kinase activity more than either doxorubicin or 8d alone. There was a positive correlation between checkpoint kinase inhibition and the improvement observed in histopathological features. Single dose treatment with doxorubicin or 8d produced S phase cell cycle arrest whereas their combination created cell cycle arrest at G2/M from 8% in case of doxorubicin to 51% in combination. Gold molecular modelling studies displayed a high correlation to the biological results.
Collapse
Affiliation(s)
- Shadia A Galal
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt.
| | - Sarah H M Khairat
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt
| | - Hamed I Ali
- Department of Pharmaceutical Sciences, Texas A&M University Irma Lerma Rangel College of Pharmacy Kingsville, TX 78363, United States; Department of Pharmaceutical Chemistry, College of Pharmacy, Helwan University, Cairo, Egypt
| | - Samia A Shouman
- Department of Cancer Biology, National Cancer Institute, Cairo University, Egypt
| | - Yasmin M Attia
- Department of Cancer Biology, National Cancer Institute, Cairo University, Egypt
| | - Mamdouh M Ali
- Department of Biochemistry, Division of Genetic Engineering and Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Abeer E Mahmoud
- Department of Biochemistry, Division of Genetic Engineering and Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Abeer H Abdel-Halim
- Department of Biochemistry, Division of Genetic Engineering and Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Amal A Fyiad
- Department of Biochemistry, Division of Genetic Engineering and Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Ashraf Tabll
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Reem El-Shenawy
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Yasmine S El Abd
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Raghda Ramdan
- Radiobiology Unit, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium; Department of Basic Medical Sciences, Physiology Group, Ghent University, Ghent, Belgium
| | - Hoda I El Diwani
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt
| |
Collapse
|
21
|
|
22
|
Niu Y, Ma F, Huang W, Fang S, Li M, Wei T, Guo L. Long non-coding RNA TUG1 is involved in cell growth and chemoresistance of small cell lung cancer by regulating LIMK2b via EZH2. Mol Cancer 2017; 16:5. [PMID: 28069000 PMCID: PMC5223434 DOI: 10.1186/s12943-016-0575-6] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 12/19/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Taurine upregulated gene1 (TUG1) as a 7.1-kb lncRNA, has been shown to play an oncogenic role in various cancers. However, the biological functions of lncRNA TUG1 in small cell lung cancer (SCLC) remain unknown. The aim of this study is to explore the roles of TUG1 in cell growth and chemoresistance of SCLC and its possible molecular mechanism. METHODS The expression of TUG1 in thirty-three cases of SCLC tissues and SCLC cell line were examined by quantitative RT-PCR (qRT-PCR). The functional roles of TUG1 in SCLC were demonstrated by CCK8 assay, colony formation assay, wound healing assay and transwell assay, flow cytometry analysis and in vivo study through siRNA or shRNA mediated knockdown. Western blot assays were used to evaluate gene and protein expression in cell lines. Chromatin immunoprecipitation (ChIP) and RNA binding protein immunoprecipitation (RIP) were performed to confirm the molecular mechanism of TUG1 involved in cell growth and chemoresistance of small cell lung cancer. RESULTS We found that TUG1 was overexpressed in SCLC tissues, and its expression was correlated with the clinical stage and the shorter survival time of SCLC patients. Moreover, downregulation of TUG1 expression could impair cell proliferation and increased cell sensitivity to anticancer drugs both in vitro and in vivo. We also discovered that TUG1 knockdown significantly promoted cell apoptosis and cell cycle arrest, and inhibited cell migration and invasion in vitro . We further demonstrated that TUG1 can regulate the expression of LIMK2b (a splice variant of LIM-kinase 2) via binding with enhancer of zeste homolog 2 (EZH2), and then promoted cell growth and chemoresistance of SCLC. CONCLUSIONS Together, these results suggested that TUG1 mediates cell growth and chemoresistance of SCLC by regulating LIMK2b via EZH2.
Collapse
Affiliation(s)
- Yuchun Niu
- Department of Pathology Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou, 510282 People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Feng Ma
- Department of Oncology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Weimei Huang
- Department of Pathology Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou, 510282 People’s Republic of China
| | - Shun Fang
- Department of Pathology Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou, 510282 People’s Republic of China
| | - Man Li
- Department of Pathology Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou, 510282 People’s Republic of China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linlang Guo
- Department of Pathology Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou, 510282 People’s Republic of China
| |
Collapse
|
23
|
Erianin induces G2/M-phase arrest, apoptosis, and autophagy via the ROS/JNK signaling pathway in human osteosarcoma cells in vitro and in vivo. Cell Death Dis 2016; 7:e2247. [PMID: 27253411 PMCID: PMC5143374 DOI: 10.1038/cddis.2016.138] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/30/2016] [Accepted: 04/21/2016] [Indexed: 02/07/2023]
Abstract
Erianin, a natural product derived from Dendrobium chrysotoxum, has exhibited potential antitumor activity in various malignancies, including hepatocarcinoma, melanoma, and promyelocytic leukemia. Here we explored the effects of erianin on osteosarcoma (OS) in vitro and in vivo and further elucidated the underlying molecule mechanisms. In this study, we found that erianin potently suppressed cell viability in various OS cell lines. Treatment with erianin induced G2/M-phase arrest, apoptosis, and autophagy in OS cells. Further studies showed that erianin-induced apoptosis and autophagy was attributed to reactive oxygen species (ROS), as N-acetyl cysteine (NAC), an ROS scavenger, attenuated them. Moreover, we found that erianin induced activation of c-Jun N-terminal kinase (JNK) signal pathway, which was also blocked by NAC. Downregulation of JNK by its specific inhibitor SP600125 could attenuate apoptosis and autophagy induced by erianin. Finally, erianin in vivo markedly reduced the growth with little organ-related toxicity. In conclusion, erianin induced cell cycle G2/M-phase arrest, apoptosis, and autophagy via the ROS/JNK signaling pathway in human OS. In light of these results, erianin may be a promising agent for anticancer therapy against OS.
Collapse
|
24
|
Hall AE, Lu WT, Godfrey JD, Antonov AV, Paicu C, Moxon S, Dalmay T, Wilczynska A, Muller PAJ, Bushell M. The cytoskeleton adaptor protein ankyrin-1 is upregulated by p53 following DNA damage and alters cell migration. Cell Death Dis 2016; 7:e2184. [PMID: 27054339 PMCID: PMC4855670 DOI: 10.1038/cddis.2016.91] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/09/2016] [Accepted: 03/15/2016] [Indexed: 12/19/2022]
Abstract
The integrity of the genome is maintained by a host of surveillance and repair mechanisms that are pivotal for cellular function. The tumour suppressor protein p53 is a major component of the DNA damage response pathway and plays a vital role in the maintenance of cell-cycle checkpoints. Here we show that a microRNA, miR-486, and its host gene ankyrin-1 (ANK1) are induced by p53 following DNA damage. Strikingly, the cytoskeleton adaptor protein ankyrin-1 was induced over 80-fold following DNA damage. ANK1 is upregulated in response to a variety of DNA damage agents in a range of cell types. We demonstrate that miR-486-5p is involved in controlling G1/S transition following DNA damage, whereas the induction of the ankyrin-1 protein alters the structure of the actin cytoskeleton and sustains limited cell migration during DNA damage. Importantly, we found that higher ANK1 expression correlates with decreased survival in cancer patients. Thus, these observations highlight ANK1 as an important effector downstream of the p53 pathway.
Collapse
Affiliation(s)
- A E Hall
- Medical Research Council (MRC), Toxicology Unit, Leicester, UK
| | - W-T Lu
- Medical Research Council (MRC), Toxicology Unit, Leicester, UK
| | - J D Godfrey
- Medical Research Council (MRC), Toxicology Unit, Leicester, UK
| | - A V Antonov
- Medical Research Council (MRC), Toxicology Unit, Leicester, UK
| | - C Paicu
- The Genome Analysis Centre, Norwich, UK.,School of Computing Sciences, University of East Anglia, Norwich, UK
| | - S Moxon
- The Genome Analysis Centre, Norwich, UK
| | - T Dalmay
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - A Wilczynska
- Medical Research Council (MRC), Toxicology Unit, Leicester, UK
| | - P A J Muller
- Medical Research Council (MRC), Toxicology Unit, Leicester, UK
| | - M Bushell
- Medical Research Council (MRC), Toxicology Unit, Leicester, UK
| |
Collapse
|
25
|
Twinstar/cofilin is required for regulation of epithelial integrity and tissue growth in Drosophila. Oncogene 2016; 35:5144-54. [DOI: 10.1038/onc.2016.46] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 01/12/2016] [Accepted: 02/08/2016] [Indexed: 12/14/2022]
|
26
|
Abstract
Cell invasion of the extracellular matrix is prerequisite to cross tissue migration of tumor cells in cancer metastasis, and vascular smooth muscle cells in atherosclerosis. The tumor suppressor p53, better known for its roles in the regulation of cell cycle and apoptosis, has ignited much interest in its function as a suppressor of cell migration and invasion. How p53 and its gain-of-function mutants regulate cell invasion remains a puzzle and a challenge for future studies. In recent years, podosomes and invadopodia have also gained center stage status as veritable apparatus specialized in cell invasion. It is not clear, however, whether p53 regulates cell invasion through podosomes and invadopodia. In this review, evidence supporting a negative role of p53 in podosomes formation in vascular smooth muscle cells will be surveyed, and signaling nodes that may mediate this regulation in other cell types will be explored.
Collapse
Affiliation(s)
- Alan S Mak
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston, ON Canada
| |
Collapse
|
27
|
Chang CY, Leu JD, Lee YJ. The actin depolymerizing factor (ADF)/cofilin signaling pathway and DNA damage responses in cancer. Int J Mol Sci 2015; 16:4095-120. [PMID: 25689427 PMCID: PMC4346946 DOI: 10.3390/ijms16024095] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/26/2015] [Accepted: 02/09/2015] [Indexed: 01/06/2023] Open
Abstract
The actin depolymerizing factor (ADF)/cofilin protein family is essential for actin dynamics, cell division, chemotaxis and tumor metastasis. Cofilin-1 (CFL-1) is a primary non-muscle isoform of the ADF/cofilin protein family accelerating the actin filamental turnover in vitro and in vivo. In response to environmental stimulation, CFL-1 enters the nucleus to regulate the actin dynamics. Although the purpose of this cytoplasm-nucleus transition remains unclear, it is speculated that the interaction between CFL-1 and DNA may influence various biological responses, including DNA damage repair. In this review, we will discuss the possible involvement of CFL-1 in DNA damage responses (DDR) induced by ionizing radiation (IR), and the implications for cancer radiotherapy.
Collapse
Affiliation(s)
- Chun-Yuan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Jyh-Der Leu
- Division of Radiation Oncology, Taipei City Hospital RenAi Branch, Taipei 106, Taiwan.
| | - Yi-Jang Lee
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
- Biophotonics & Molecular Imaging Research Center (BMIRC), National Yang-Ming University, Taipei 112, Taiwan.
| |
Collapse
|
28
|
Lourenço FC, Munro J, Brown J, Cordero J, Stefanatos R, Strathdee K, Orange C, Feller SM, Sansom OJ, Vidal M, Murray GI, Olson MF. Reduced LIMK2 expression in colorectal cancer reflects its role in limiting stem cell proliferation. Gut 2014; 63:480-93. [PMID: 23585469 PMCID: PMC3932979 DOI: 10.1136/gutjnl-2012-303883] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 02/12/2013] [Accepted: 03/24/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Colorectal cancer (CRC) is a major contributor to cancer mortality and morbidity. LIM kinase 2 (LIMK2) promotes tumour cell invasion and metastasis. The objectives of this study were to determine how LIMK2 expression is associated with CRC progression and patient outcome, and to use genetically modified Drosophila and mice to determine how LIMK2 deletion affects gastrointestinal stem cell regulation and tumour development. DESIGN LIMK2 expression and activity were measured by immunostaining tumours from CRC-prone mice, human CRC cell lines and 650 human tumours. LIMK knockdown in Drosophila or Limk2 deletion in mice allowed for assessment of their contributions to gastrointestinal stem cell homeostasis and tumour development. RESULTS LIMK2 expression was reduced in intestinal tumours of cancer-prone mice, as well as in human CRC cell lines and tumours. Reduced LIMK2 expression and substrate phosphorylation were associated with shorter patient survival. Genetic analysis in Drosophila midgut and intestinal epithelial cells isolated from genetically modified mice revealed a conserved role for LIMK2 in constraining gastrointestinal stem cell proliferation. Limk2 deletion increased colon tumour size in a colitis-associated colorectal mouse cancer model. CONCLUSIONS This study revealed that LIMK2 expression and activity progressively decrease with advancing stage, and supports the hypothesis that there is selective pressure for reduced LIMK2 expression in CRC to relieve negative constraints imposed upon gastrointestinal stem cells.
Collapse
Affiliation(s)
| | - June Munro
- Beatson Institute for Cancer Research, Glasgow, UK
| | | | | | | | | | - Clare Orange
- Department of Pathology, Division of Cancer Sciences and Molecular Pathology, Western Infirmary, Glasgow, UK
| | - Stephan M Feller
- Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Marcos Vidal
- Beatson Institute for Cancer Research, Glasgow, UK
| | - Graeme I Murray
- Department of Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
29
|
Kang MS, Yu SL, Kim HY, Gorospe CM, Choi BH, Lee SH, Lee SK. Yeast RAD2, a homolog of human XPG, plays a key role in the regulation of the cell cycle and actin dynamics. Biol Open 2014; 3:29-41. [PMID: 24326185 PMCID: PMC3892158 DOI: 10.1242/bio.20136403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mutations in the human XPG gene cause Cockayne syndrome (CS) and xeroderma pigmentosum (XP). Transcription defects have been suggested as the fundamental cause of CS; however, defining CS as a transcription syndrome is inconclusive. In particular, the function of XPG in transcription has not been clearly demonstrated. Here, we provide evidence for the involvement of RAD2, the Saccharomyces cerevisiae counterpart of XPG, in cell cycle regulation and efficient actin assembly following ultraviolet irradiation. RAD2 C-terminal deletion, which resembles the XPG mutation found in XPG/CS cells, caused cell growth arrest, the cell cycle stalling, a defective α-factor response, shortened lifespan, cell polarity defect, and misregulated actin-dynamics after DNA damage. Overexpression of the C-terminal 65 amino acids of Rad2p was sufficient to induce hyper-cell polarization. In addition, RAD2 genetically interacts with TPM1 during cell polarization. These results provide insights into the role of RAD2 in post-UV irradiation cell cycle regulation and actin assembly, which may be an underlying cause of XPG/CS.
Collapse
Affiliation(s)
- Mi-Sun Kang
- Department of Pharmacology, College of Medicine, Inha University, Incheon, Korea 400-712
| | | | | | | | | | | | | |
Collapse
|
30
|
Gamell C, Schofield AV, Suryadinata R, Sarcevic B, Bernard O. LIMK2 mediates resistance to chemotherapeutic drugs in neuroblastoma cells through regulation of drug-induced cell cycle arrest. PLoS One 2013; 8:e72850. [PMID: 23991158 PMCID: PMC3749167 DOI: 10.1371/journal.pone.0072850] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 07/16/2013] [Indexed: 01/12/2023] Open
Abstract
Drug resistance is a major obstacle for the successful treatment of many malignancies, including neuroblastoma, the most common extracranial solid tumor in childhood. Therefore, current attempts to improve the survival of neuroblastoma patients, as well as those with other cancers, largely depend on strategies to counter cancer cell drug resistance; hence, it is critical to understand the molecular mechanisms that mediate resistance to chemotherapeutics. The levels of LIM-kinase 2 (LIMK2) are increased in neuroblastoma cells selected for their resistance to microtubule-targeted drugs, suggesting that LIMK2 might be a possible target to overcome drug resistance. Here, we report that depletion of LIMK2 sensitizes SHEP neuroblastoma cells to several microtubule-targeted drugs, and that this increased sensitivity correlates with enhanced cell cycle arrest and apoptosis. Furthermore, we show that LIMK2 modulates microtubule acetylation and the levels of tubulin Polymerization Promoting Protein 1 (TPPP1), suggesting that LIMK2 may participate in the mitotic block induced by microtubule-targeted drugs through regulation of the microtubule network. Moreover, LIMK2-depleted cells also show an increased sensitivity to certain DNA-damage agents, suggesting that LIMK2 might act as a general pro-survival factor. Our results highlight the exciting possibility of combining specific LIMK2 inhibitors with anticancer drugs in the treatment of multi-drug resistant cancers.
Collapse
Affiliation(s)
- Cristina Gamell
- Cytoskeleton and Cancer Unit, St. Vincent’s Institute of Medical Research, Melbourne, Victoria, Australia
| | - Alice V. Schofield
- Cytoskeleton and Cancer Unit, St. Vincent’s Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medicine at St. Vincent’s Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Randy Suryadinata
- Cell Cycle and Cancer Unit, St. Vincent’s Institute of Medical Research, Melbourne, Victoria, Australia
| | - Boris Sarcevic
- Department of Medicine at St. Vincent’s Hospital, The University of Melbourne, Melbourne, Victoria, Australia
- Cell Cycle and Cancer Unit, St. Vincent’s Institute of Medical Research, Melbourne, Victoria, Australia
| | - Ora Bernard
- Cytoskeleton and Cancer Unit, St. Vincent’s Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medicine at St. Vincent’s Hospital, The University of Melbourne, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
31
|
Huang L, Kuwahara I, Matsumoto K. EWS represses cofilin 1 expression by inducing nuclear retention of cofilin 1 mRNA. Oncogene 2013; 33:2995-3003. [PMID: 23831569 DOI: 10.1038/onc.2013.255] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 04/05/2013] [Accepted: 05/06/2013] [Indexed: 12/15/2022]
Abstract
In Ewing's sarcoma family tumors (ESFTs), the proto-oncogene EWS that encodes an RNA-binding protein is fused by chromosomal translocation to the gene encoding one of the E-twenty six (ETS) family of transcription factors, most commonly friend leukemia virus integration 1 (FLI-1). Although EWS/FLI-1 chimeric proteins are necessary for carcinogenesis, additional events seem to be required for transformation to occur. We have previously reported that a protein product of an EWS mRNA target, whose expression is negatively regulated by EWS but not by EWS/FLI-1, contributes to ESFT development. However, the mechanism by which EWS represses protein expression remains to be elucidated. Here, we report that overexpression of full-length EWS repressed protein expression and induced nuclear retention of reporter mRNAs in a tethering assay. In contrast, when a mutant lacking the EWS C-terminal nuclear localization signal (classified as a PY-NLS) was expressed, reporter protein expression was upregulated, and the number of cells exporting reporter mRNA to the cytoplasm increased. EWS binds to the 3'-untranslated region in another mRNA target, cofilin 1 (CFL1), and negatively regulates the expression of CFL1. Overexpression of EWS induced nuclear retention of CFL1 mRNA. Furthermore, ESFT cell proliferation and metastatic potential were suppressed by small interfering RNA-mediated CFL1 knockdown. Together, our findings suggest that EWS induces nuclear retention of CFL1 mRNA, thereby suppressing expression of CFL1, and that CFL1 promotes development of ESFT. Targeting CFL1 might therefore provide another novel approach for treatment of this aggressive disease.
Collapse
Affiliation(s)
- L Huang
- 1] Molecular Entomology Laboratory, RIKEN, Wako, Japan [2] Department of Pathophysiology, Dalian Medical University, Dalian, China
| | - I Kuwahara
- Molecular Entomology Laboratory, RIKEN, Wako, Japan
| | - K Matsumoto
- 1] Molecular Entomology Laboratory, RIKEN, Wako, Japan [2] PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| |
Collapse
|
32
|
Sun C, Zheng J, Cheng S, Feng D, He J. EBP50 phosphorylation by Cdc2/Cyclin B kinase affects actin cytoskeleton reorganization and regulates functions of human breast cancer cell line MDA-MB-231. Mol Cells 2013; 36:47-54. [PMID: 23775624 PMCID: PMC3887931 DOI: 10.1007/s10059-013-0014-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 04/29/2013] [Accepted: 05/03/2013] [Indexed: 10/26/2022] Open
Abstract
The actin cytoskeleton plays an important role in cell shape determination, adhesion and cell cycle progression. Ezrinradixin-moesin (ERM)-binding phosphoprotein 50 (EBP50), also known as Na(+)-H(+) exchanger regulatory factor 1 (NHERF1), associates with actin cytoskeleton and is related to cell cycle progression. Its Ser279 and Ser301 residues are phosphorylated by cyclin-dependent kinase 2 (cdc2)/cyclin B during the mitosis phase. However, the biological significance of EBP50 phosphorylation mediated by cdc2/cyclin B is not clear. In the present study, MDA-MB-231 cells with low levels of endogenous EBP50 protein were stably transfected with constructs of EBP50 wild type (WT), phosphodeficient (serine 279 and serine 301 mutated to alanine-S279A/S301A) or phospho-mimetic (serine 279 and serine 301 mutated to aspartic acid-S279D/S301D) mutants. Subsequently, multiple phenotypes of these cells were characterized. Failure of cdc2/cyclin B-mediated EBP50 phosphorylation in cells expressing S279A/S301A (AA cells) significantly increased F-actin content, enhanced the adherence of cells to the extracellular matrix, altered cell morphology and caused defects in cytokinesis, as reflected in the formation of giant cells with heteroploid DNA and multinucleation or giant nuclei. Furthermore, knockdown of EBP50 expression in AA cells rescued cell defects such as the cytokinesis failure and abnormal cell morphology. EBP50 S279A/ S301A had a weaker binding affinity with actin than EBP50 S279D/S301D, which might explain the increase of F-actin content in the AA cells. The present results suggest that cdc2/cyclin B-mediated EBP50 phosphorylation may play a role in the regulation of various cell functions by affecting actin cytoskeleton reorganization.
Collapse
Affiliation(s)
- Chaoyuan Sun
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069,
China
| | - Junfang Zheng
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069,
China
| | - Shan Cheng
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069,
China
| | - Duiping Feng
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001,
China
| | - Junqi He
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069,
China
| |
Collapse
|
33
|
Schofield AV, Bernard O. Rho-associated coiled-coil kinase (ROCK) signaling and disease. Crit Rev Biochem Mol Biol 2013; 48:301-16. [PMID: 23601011 DOI: 10.3109/10409238.2013.786671] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The small Rho GTPase family of proteins, encompassing the three major G-protein classes Rho, Rac and cell division control protein 42, are key mitogenic signaling molecules that regulate multiple cancer-associated cellular phenotypes including cell proliferation and motility. These proteins are known for their role in the regulation of actin cytoskeletal dynamics, which is achieved through modulating the activity of their downstream effector molecules. The Rho-associated coiled-coil kinase 1 and 2 (ROCK1 and ROCK2) proteins were the first discovered Rho effectors that were primarily established as players in RhoA-mediated stress fiber formation and focal adhesion assembly. It has since been discovered that the ROCK kinases actively phosphorylate a large cohort of actin-binding proteins and intermediate filament proteins to modulate their functions. It is well established that global cellular morphology, as modulated by the three cytoskeletal networks: actin filaments, intermediate filaments and microtubules, is regulated by a variety of accessory proteins whose activities are dependent on their phosphorylation by the Rho-kinases. As a consequence, they regulate many key cellular functions associated with malignancy, including cell proliferation, motility and viability. In this current review, we focus on the role of the ROCK-signaling pathways in disease including cancer.
Collapse
Affiliation(s)
- Alice V Schofield
- St Vincent's Institute of Medical Research, Cytoskeleton and Cancer Unit and Department of Medicine, St Vincent's Hospital, University of Melbourne, Victoria 3065, Australia
| | | |
Collapse
|
34
|
Leu JD, Chiu YW, Lo CC, Chiang PH, Chiu SJ, Tsai CH, Hwang JJ, Chen RC, Gorbunova V, Lee YJ. Enhanced cellular radiosensitivity induced by cofilin-1 over-expression is associated with reduced DNA repair capacity. Int J Radiat Biol 2013; 89:433-44. [PMID: 23362981 DOI: 10.3109/09553002.2013.767992] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE A previous report has indicated that over-expression of cofilin-1 (CFL-1), a member of the actin depolymerizing factor (ADF)/cofilin protein family, enhances cellular radiosensitivity. This study explores the involvement of various DNA damage responses and repair systems in the enhanced cellular radiosensitivity as well as assessing the role of CFL-1 phosphorylation in radiosensitivity. MATERIALS AND METHODS Human non-small lung cancer H1299 cells harboring a tet-on gene expression system were used to induce exogenous expression of wild-type CFL-1. Colony formation assays were used to determine cell survival after γ-ray exposure. DNA damage levels were determined by Comet assay. DNA repair capacity was assessed by fluorescence-based DNA repair analysis and antibody detection of various repair proteins. The effects of CFL-1 phosphorylation on radiation responses were explored using two mutant CFL-1 proteins, S3D and S3A. Finally, endogenous CFL-1 phosphorylation levels were investigated using latrunculin A (LA), cytochalasin B (CB) and Y27632. RESULTS When phosphorylatable CFL-1 was expressed, radiosensitivity was enhanced after exposure to γ-rays and this was accompanied by DNA damage. Phosphorylated histone H2AX (γ-H2AX) and p53-binding protein-1 (53BP1) foci, as well as Chk1/2 phosphorylation, were apparently suppressed, although ataxia telangiectasia mutated (ATM) kinase activation was apparently unaffected. In addition, two radiation-induced double-strand break (DSB) repair systems, namely homologous recombination repair (HRR) and non-homologous end joining (NHEJ), were suppressed. Moreover, over-expression of CFL-1 S3D and CFL-1 S3A both enhanced radiosensitivity. However, enhanced radiosensitivity and reduced γ-H2AX expression were only detected in cells treated with LA which increased endogenous phospho-CFL-1, and not in cells treated with Y27632, which dephosphorylates CFL-1. CONCLUSION CFL-1 over-expression enhances radiosensitivity and this is associated with reduced DNA repair capacity. Although phosphorylated CFL-1 seems to be involved in radiosensitivity, further studies are required to address the importance of CFL-1 activity to the regulation of radiosensitivity.
Collapse
Affiliation(s)
- Jyh-Der Leu
- Division of Radiation Oncology, Taipei City Hospital RenAi Branch , Taipei
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Signaling through the Rho family of small GTPases regulates a variety of cellular processes via changes in the actin cytoskeleton. Here we discuss recent findings that show the transcription factor p53 regulates the expression of several Rho pathway signaling molecules, and how mutation of p53 in cancer dramatically alters signaling output through this pathway.
Collapse
Affiliation(s)
- Daniel R Croft
- The Beatson Institute for Cancer Research, Garscube Estate; Glasgow, UK
| | | |
Collapse
|
36
|
Matsuse M, Sasaki K, Nishihara E, Minami S, Hayashida C, Kondo H, Suzuki K, Saenko V, Yoshiura KI, Mitsutake N, Yamashita S. Copy number alteration and uniparental disomy analysis categorizes Japanese papillary thyroid carcinomas into distinct groups. PLoS One 2012; 7:e36063. [PMID: 22558328 PMCID: PMC3340412 DOI: 10.1371/journal.pone.0036063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Accepted: 03/30/2012] [Indexed: 02/03/2023] Open
Abstract
The aim of the present study was to investigate chromosomal aberrations in sporadic Japanese papillary thyroid carcinomas (PTCs), concomitant with the analysis of oncogene mutational status. Twenty-five PTCs (11 with BRAF(V600E), 4 with RET/PTC1, and 10 without mutation in HRAS, KRAS, NRAS, BRAF, RET/PTC1, or RET/PTC3) were analyzed using Genome-Wide Human SNP Array 6.0 which allows us to detect copy number alteration (CNA) and uniparental disomy (UPD), also referred to as copy neutral loss of heterozygosity, in a single experiment. The Japanese PTCs showed relatively stable karyotypes. Seven cases (28%) showed CNA(s), and 6 (24%) showed UPD(s). Interestingly, CNA and UPD were rarely overlapped in the same tumor; the only one advanced case showed both CNA and UPD with a highly complex karyotype. Thirteen (52%) showed neither CNA nor UPD. Regarding CNA, deletions tended to be more frequent than amplifications. The most frequent and recurrent region was the deletion in chromosome 22; however, it was found in only 4 cases (16%). The degree of genomic instability did not depend on the oncogene status. However, in oncogene-positive cases (BRAF(V600E) and RET/PTC1), tumors with CNA/UPD were less frequent (5/15, 33%), whereas tumors with CNA/UPD were more frequent in oncogene-negative cases (7/10, 70%), suggesting that chromosomal aberrations may play a role in the development of PTC, especially in oncogene-negative tumors. These data suggest that Japanese PTCs may be classified into three distinct groups: CNA(+), UPD(+), and no chromosomal aberrations. BRAF(V600E) mutational status did not correlate with any parameters of chromosomal defects.
Collapse
Affiliation(s)
- Michiko Matsuse
- Department of Radiation Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Kensaku Sasaki
- Department of Human Genetics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Eijun Nishihara
- Department of Internal Medicine, Kuma Hospital, Kobe, Hyogo, Japan
| | - Shigeki Minami
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Chisa Hayashida
- Department of Human Genetics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Hisayoshi Kondo
- Division of Scientific Data Registry, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Keiji Suzuki
- Department of Radiation Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Vladimir Saenko
- Department of Health Risk Control, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Koh-ichiro Yoshiura
- Department of Human Genetics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | - Norisato Mitsutake
- Department of Radiation Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
- Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (NRGIC), Nagasaki, Nagasaki, Japan
| | - Shunichi Yamashita
- Department of Radiation Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
- Department of Health Risk Control, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| |
Collapse
|
37
|
Bensimon A, Aebersold R, Shiloh Y. Beyond ATM: the protein kinase landscape of the DNA damage response. FEBS Lett 2011; 585:1625-39. [PMID: 21570395 DOI: 10.1016/j.febslet.2011.05.013] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 05/04/2011] [Accepted: 05/04/2011] [Indexed: 01/18/2023]
Abstract
The DNA of all organisms is constantly subjected to damaging agents, both exogenous and endogenous. One extremely harmful lesion is the double-strand break (DSB), which activates a massive signaling network - the DNA damage response (DDR). The chief activator of the DSB response is the ATM protein kinase, which phosphorylates numerous key players in its various branches. Recent phosphoproteomic screens have extended the scope of damage-induced phosphorylations beyond the direct ATM substrates. We review the evidence for the involvement of numerous other protein kinases in the DDR, obtained from documentation of specific pathways as well as high-throughput screens. The emerging picture of the protein phosphorylation landscape in the DDR broadens the current view on the role of this protein modification in the maintenance of genomic stability. Extensive cross-talk between many of these protein kinases forms an interlaced signaling network that spans numerous cellular processes. Versatile protein kinases in this network affect pathways that are different from those they have been identified with to date. The DDR appears to be one of the most extensive signaling responses to cellular stimuli.
Collapse
Affiliation(s)
- Ariel Bensimon
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | | | | |
Collapse
|
38
|
Croft DR, Crighton D, Samuel MS, Lourenco FC, Munro J, Wood J, Bensaad K, Vousden KH, Sansom OJ, Ryan KM, Olson MF. p53-mediated transcriptional regulation and activation of the actin cytoskeleton regulatory RhoC to LIMK2 signaling pathway promotes cell survival. Cell Res 2011; 21:666-82. [PMID: 21079653 PMCID: PMC3145139 DOI: 10.1038/cr.2010.154] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Revised: 09/20/2010] [Accepted: 09/26/2010] [Indexed: 12/28/2022] Open
Abstract
The central arbiter of cell fate in response to DNA damage is p53, which regulates the expression of genes involved in cell cycle arrest, survival and apoptosis. Although many responses initiated by DNA damage have been characterized, the role of actin cytoskeleton regulators is largely unknown. We now show that RhoC and LIM kinase 2 (LIMK2) are direct p53 target genes induced by genotoxic agents. Although RhoC and LIMK2 have well-established roles in actin cytoskeleton regulation, our results indicate that activation of LIMK2 also has a pro-survival function following DNA damage. LIMK inhibition by siRNA-mediated knockdown or selective pharmacological blockade sensitized cells to radio- or chemotherapy, such that treatments that were sub-lethal when administered singly resulted in cell death when combined with LIMK inhibition. Our findings suggest that combining LIMK inhibitors with genotoxic therapies could be more efficacious than single-agent administration, and highlight a novel connection between actin cytoskeleton regulators and DNA damage-induced cell survival mechanisms.
Collapse
Affiliation(s)
- Daniel R Croft
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Diane Crighton
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Current address: Cancer Research Technology, The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Michael S Samuel
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Filipe C Lourenco
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - June Munro
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jenifer Wood
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karim Bensaad
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Current address: Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Karen H Vousden
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Owen J Sansom
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Kevin M Ryan
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Michael F Olson
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
39
|
Gutierrez O, Berciano MT, Lafarga M, Fernandez-Luna JL. A novel pathway of TEF regulation mediated by microRNA-125b contributes to the control of actin distribution and cell shape in fibroblasts. PLoS One 2011; 6:e17169. [PMID: 21347262 PMCID: PMC3037971 DOI: 10.1371/journal.pone.0017169] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 01/24/2011] [Indexed: 02/02/2023] Open
Abstract
Background Thyrotroph embryonic factor (TEF), a member of the PAR bZIP family of transcriptional regulators, has been involved in neurotransmitter homeostasis, amino acid metabolism, and regulation of apoptotic proteins. In spite of its relevance, nothing is known about the regulation of TEF. Principal Findings p53-dependent genotoxic agents have been shown to be much more harmful for PAR bZIP-deficient mice as compared to wild type animals. Here we demonstrate that TEF expression is controlled by p53 through upregulation of microRNA-125b, as determined by both regulating the activity of p53 and transfecting cells with microRNA-125b precursors. We also describe a novel role for TEF in controlling actin distribution and cell shape in mouse fibroblasts. Lack of TEF is accompanied by dramatic increase of cell area and decrease of elongation (bipolarity) and dispersion (multipolarity). Staining of actin cytoskeleton also showed that TEF (−/−) cells are characterized by appearance of circumferential actin bundles and disappearance of straight fibers. Interestingly, transfection of TEF (−/−) fibroblasts with TEF induced a wild type-like phenotype. Consistent with our previous findings, transfection of wild type fibroblasts with miR-125b promoted a TEF (−/−)-like phenotype, and a similar but weaker effect was observed following exogenous expression of p53. Conclusions/Significance These findings provide the first evidence of TEF regulation, through a miR-125b-mediated pathway, and describes a novel role of TEF in the maintenance of cell shape in fibroblasts.
Collapse
Affiliation(s)
- Olga Gutierrez
- Unidad de Genetica Molecular, Hospital Universitario Marques de Valdecilla, Instituto de Formacion e Investigacion Marques de Valdecilla, Servicio Cantabro de Salud, Santander, Spain
| | - Maria T. Berciano
- Departamento de Anatomia y Biologia Celular, Universidad de Cantabria, Santander, Spain
| | - Miguel Lafarga
- Departamento de Anatomia y Biologia Celular, Universidad de Cantabria, Santander, Spain
| | - Jose L. Fernandez-Luna
- Unidad de Genetica Molecular, Hospital Universitario Marques de Valdecilla, Instituto de Formacion e Investigacion Marques de Valdecilla, Servicio Cantabro de Salud, Santander, Spain
- * E-mail:
| |
Collapse
|
40
|
Oleinik NV, Krupenko NI, Krupenko SA. ALDH1L1 inhibits cell motility via dephosphorylation of cofilin by PP1 and PP2A. Oncogene 2010; 29:6233-44. [PMID: 20729910 PMCID: PMC2992098 DOI: 10.1038/onc.2010.356] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Here we report that ALDH1L1 (FDH, a folate enzyme with tumor suppressor-like properties) inhibits cell motility. The underlying mechanism involves F-actin stabilization, re-distribution of cytoplasmic actin towards strong preponderance of filamentous actin, and formation of actin stress fibers. A549 cells expressing FDH demonstrated a much slower recovery of GFP-actin fluorescence in a FRAP assay, as well as an increase in G-actin polymerization and a decrease in F-actin depolymerization rates in pyren-actin fluorescence assays indicating the inhibition of actin dynamics. These effects were associated with robust dephosphorylation of the actin depolymerizing factor cofilin by PP1 and PP2A serine/threonine protein phosphatases but not the cofilin-specific phosphatases slingshot and chronophin. In fact, the PP1/PP2A inhibitor calyculin prevented cofilin dephosphorylation and restored motility. Inhibition of FDH-induced apoptosis by the JNK inhibitor SP600125 or the pan-caspase inhibitor zVAD-fmk did not restore motility or levels of phospho-cofilin, indicating that the observed effects are independent from FDH function in apoptosis. Interestingly, cofilin siRNA or expression of phosphorylation-deficient S3A cofilin mutant resulted in a decrease of G-actin and the actin stress fiber formation, the effects seen upon FDH expression. In contrast, the expression of S3D mutant, mimicking constitutive phosphorylation, prevented these effects further supporting the cofilin-dependent mechanism. Dephosphorylation of cofilin and inhibition of motility in response to FDH can be also prevented by the increased folate in media. Furthermore, folate depletion itself, in the absence of FDH, resulted in cofilin dephosphorylation and inhibition of motility in several cell lines. Our experiments showed that these effects were folate-specific and not a general response to nutrient starvation. Overall, this study demonstrates the presence of distinct intracellular signaling pathways regulating motility in response to folate status and points toward mechanisms involving folates in promoting a malignant phenotype.
Collapse
Affiliation(s)
- N V Oleinik
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | | | | |
Collapse
|