1
|
Shan S, Su M, Wang H, Guo F, Li Y, Zhou Y, Liu H, Du L, Zhang J, Qiu J, DiSanto ME, Guo Y, Zhang X. Y-27632 targeting ROCK1&2 modulates cell growth, fibrosis and epithelial-mesenchymal transition in hyperplastic prostate by inhibiting β-catenin pathway. MOLECULAR BIOMEDICINE 2024; 5:52. [PMID: 39455522 PMCID: PMC11511810 DOI: 10.1186/s43556-024-00216-9] [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: 05/25/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) is a prevalent condition affecting the male urinary system, with its molecular mechanisms of pathogenesis remaining unclear. Y-27632, a non-isoform-selective Rho kinase inhibitor, has shown therapeutic potential in various diseases but its effects on static factors and fibrosis in BPH remain unexplored. This study investigated human prostate tissues, human prostate cell lines, and BPH rat model using immunofluorescence, flow cytometry, quantitative reverse transcription polymerase chain reaction, western blotting, and cell counting kit-8. ROCK1 and ROCK2 were significantly up-regulated in BPH tissues, correlating with clinical parameters. Y-27632 targeted the inhibition of ROCK1 & ROCK2 expression and inhibited cell proliferation, fibrosis, epithelial-mesenchymal transition (EMT), while induced cell apoptosis in a dose-dependent manner. Moreover, knockdown of either ROCK isoform inhibited fibrosis and EMT, induced apoptosis, while ROCK overexpression had the opposite effects. ROCK downregulation inhibited the β-catenin signaling pathway (such as C-MYC, Snail and Survivin) and decreased β-catenin protein stability, while inhibiting TGF-β/Smad2/3 signaling. At the in vivo level, Y-27632 reversed prostatic hyperplasia and fibrosis in BPH model rats to some extent. Our study sheds light on the therapeutic potential of Y-27632 in regulating prostate cell growth, fibrosis and EMT, and demonstrates for the first time the regulatory effect of ROCK isoforms on prostate cells, providing the basis for future research of ROCK isoform-selective inhibitors.
Collapse
Affiliation(s)
- Shidong Shan
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Renal Transplatation, Guangdong Provincial People' Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Min Su
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hejin Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Feng Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Li
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yongying Zhou
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Huan Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lu Du
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junchao Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jizhang Qiu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Michael E DiSanto
- Department of Surgery and Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Yuming Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xinhua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Cao Z, Wei X, Xing S, Zhang J, Wang S, Yue L, Zhang J, Jiang N, Zhai X. Discovery of novel pyrazolo[1,5-a]pyrimidine derivatives as selective ROCK2 inhibitors with anti-breast cancer migration and invasion activities. Bioorg Chem 2024; 151:107675. [PMID: 39126868 DOI: 10.1016/j.bioorg.2024.107675] [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: 06/14/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
Rho-associated coiled-coil kinase (ROCK) is involved in multiple cellular activities regulating the actin cytoskeleton, such as cell morphology, adhesion, and migration. The inhibition of ROCK is a feasible strategy to suppress breast cancer metastasis. Herein, based on Belumosudil, a series of pyrazolo[1,5-a]pyrimidine derivatives as selective ROCK2 inhibitors were designed and synthesized. Through systematic investigation of SARs, the piperazine analog 7u was identified with optimum ROCK2 inhibitory activity (IC50 = 36.8 nM) and excellent selectivity over the isoform protein ROCK1 (>250-fold). Intriguingly, upon treatment with 7u, the arrangement of the MDA-MB-231 cytoskeleton was affected accompanied by the alteration of morphology. Furthermore, cell scratch and transwell assays indicated that 7u inhibited MDA-MB-231 cell migration and invasion in a dose-dependent manner. Ultimately, the binding model of 7u with ROCK2 well accounted for the superior activities of 7u as a promising ROCK2 inhibitor with the potential application in breast cancer metastasis treatment.
Collapse
Affiliation(s)
- Zhi Cao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiujian Wei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuming Xing
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiahao Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuqi Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lingfeng Yue
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiahe Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Nan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
3
|
Xu X, Yao L. Recent advances in the development of Rho kinase inhibitors (2015-2021). Med Res Rev 2024; 44:406-421. [PMID: 37265266 DOI: 10.1002/med.21980] [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: 12/23/2021] [Revised: 01/27/2023] [Accepted: 05/23/2023] [Indexed: 06/03/2023]
Abstract
Rho-associated coiled-coil kinases (ROCKs) are key downstream effectors of small GTPases. ROCK plays a central role in diverse cellular events with accumulating evidence supporting the concept that ROCK is important in tumor development and progression. Numerous ROCK inhibitors have been investigated for their therapeutic potential in the treatment of cancers. In this article, we review recent research progress on ROCK inhibitors, especially those with potential for the treatment of cancers, reported in the literature from 2015 to 2021. Most ROCK inhibitors show potent in vitro and in vivo antitumor activities and have potential in the treatment of cancers.
Collapse
Affiliation(s)
- Xiangrong Xu
- Yantai University Hospital, Yantai University, Yantai, China
| | - Lei Yao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
4
|
Wu Q, Leng X, Ma X, Xu Q, Ni R, Pan Y, Jin Z, Kalim M. Triptolide Reduces MDA-MB-231 Cell Metastasis by Attenuating Epithelial-Mesenchymal Transition through the ROCK/PTEN/Akt Axis. Chem Biodivers 2023; 20:e202300399. [PMID: 37910661 DOI: 10.1002/cbdv.202300399] [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: 03/22/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/03/2023]
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous and invasive subtype of breast cancer. The prognosis of TNBC is poor because of its high distant metastasis rate. Triptolide is a type of diterpene trioxide natural compound with potential anti-tumor activities. This study explored the metastatic inhibitory effect of triptolide on MDA-MB-231 cells and its underlying mechanism. Triptolide suppressed cell proliferation and induced cell apoptosis in a time- and dose-dependent manner. Low doses of triptolide (0-8 nM) reduced the migration and invasion capabilities of MDA-MB-231 cells. Triptolide decreased ROCK1, p-Akt, N-cadherin, vimentin and MMP-9 expressions, but increased PTEN and E-cadherin expressions on protein and mRNA levels. Furthermore, the down-regulation of ROCK1 expression in MDA-MB-231 cells after being treated by triptolide could be rescued by ROCK1 specific inhibitor Y27632. Molecular docking showed that triptolide and Y27632 shared the same active center of ROCK1 protein. This article's findings taken together showed that ROCK1 is the primary target of triptolide, which can cause cell apoptosis and inhibit the epithelial-mesenchymal transition of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Qinhang Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Xuejiao Leng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Xuelin Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Qixuan Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Ruijun Ni
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Yang Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Zhuolin Jin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Muhammad Kalim
- Department of Microbiology and Immunology, Wake Forest University, Winston-Salem, NC, 27101, USA
| |
Collapse
|
5
|
Wu Q, Ma X, Jin Z, Ni R, Pan Y, Yang G. Zhuidu Formula suppresses the migratory and invasive properties of triple-negative breast cancer cells via dual signaling pathways of RhoA/ROCK and CDC42/MRCK. JOURNAL OF ETHNOPHARMACOLOGY 2023; 315:116644. [PMID: 37196814 DOI: 10.1016/j.jep.2023.116644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhuidu Formula (ZDF) is composed of triptolide, cinobufagin and paclitaxel, which are the active ingredients of Tripterygium wilfordii Hook. F, dried toad skin and Taxus wallichiana var. chinensis (Pilg) Florin, respectively. Modern pharmacological studies show that triptolide, cinobufagin, and paclitaxel are well-known natural compounds that exert anti-tumor effects by interfering with DNA synthesis, inducing tumor cell apoptosis, and inhibiting the dynamic balance of the tubulin. However, the mechanism by which the three compounds inhibit triple-negative breast cancer (TNBC) metastasis is unknown. OBJECTIVE The objective of this investigation was to examine the inhibitory essences of ZDF on the metastasis of TNBC and elucidate its potential mechanism. MATERIALS AND METHODS Cell viability of triptolide (TPL), cinobufagin (CBF), and paclitaxel (PTX) on MDA-MB-231 cells was assessed employing a CCK-8 assay. The drug interactions of the three drugs on MDA-MB-231 cells were determined in vitro utilizing the Chou-Talalay method. MDA-MB-231 cells were identified for migration, invasion and adhesion in vitro through the implementation of the scratch assay, transwell assay and adhesion assay, respectively. The formation of cytoskeleton protein F-actin was detected by immunofluorescence assay. The expressions of MMP-2 and MMP-9 in the supernatant of the cells were determined by ELISA analysis. The Western blot and RT-qPCR were employed to explore the protein expressions associated with the dual signaling pathways of RhoA/ROCK and CDC42/MRCK. The anti-tumor efficacy of ZDF in vivo and its preliminary mechanism were investigated in the mouse 4T1 TNBC model. RESULTS The results demonstrated that ZDF could significantly reduce the viability of the MDA-MB-231 cell, and the combination index (CI) values of actual compatibility experimental points were all less than 1, demonstrating a favorable synergistic compatibility relationship. It was found that ZDF reduces RhoA/ROCK and CDC42/MRCK dual signaling pathways, which are responsible for MDA-MB-231cell migration, invasion, and adhesion. Additionally, there has been a significant reduction in the manifestation of cytoskeleton-related proteins. Furthermore, the expression levels of RhoA, CDC42, ROCK2, and MRCKβ mRNA and protein were down-regulated. ZDF significantly decreased the protein expressions of vimentin, cytokeratin-8, Arp2 and N-WASP, and inhibited actin polymerization and actomyosin contraction. Furthermore, MMP-2 and MMP-9 levels in the high-dose ZDF group were decreased by 30% and 26%, respectively. ZDF significantly reduced the tumor volume and protein expressions of ROCK2 and MRCKβ in tumor tissues without eliciting any perceptible alterations in the physical mass of the mice, and the reduction was more pronounced than that of the BDP5290 treated group. CONCLUSION The current investigation demonstrates that ZDF exhibits a proficient inhibitory impact on TNBC metastasis by regulating cytoskeletal proteins through the dual signaling pathways of RhoA/ROCK and CDC42/MRCK. Furthermore, the findings indicate that ZDF has significant anti-tumorigenic and anti-metastatic characteristics in breast cancer animal models.
Collapse
Affiliation(s)
- Qinhang Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Xuelin Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Zhuolin Jin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Ruijun Ni
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Yang Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| | - Guangming Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| |
Collapse
|
6
|
Kale VP, Hengst JA, Sharma AK, Golla U, Dovat S, Amin SG, Yun JK, Desai DH. Characterization of Anticancer Effects of the Analogs of DJ4, a Novel Selective Inhibitor of ROCK and MRCK Kinases. Pharmaceuticals (Basel) 2023; 16:1060. [PMID: 37630974 PMCID: PMC10458458 DOI: 10.3390/ph16081060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/17/2023] [Accepted: 07/22/2023] [Indexed: 08/27/2023] Open
Abstract
The Rho associated coiled-coil containing protein kinase (ROCK1 and ROCK2) and myotonic dystrophy-related Cdc-42 binding kinases (MRCKα and MRCKβ) are critical regulators of cell proliferation and cell plasticity, a process intimately involved in cancer cell migration and invasion. Previously, we reported the discovery of a novel small molecule (DJ4) selective multi-kinase inhibitor of ROCK1/2 and MRCKα/β. Herein, we further characterized the anti-proliferative and apoptotic effects of DJ4 in non-small cell lung cancer and triple-negative breast cancer cells. To further optimize the ROCK/MRCK inhibitory potency of DJ4, we generated a library of 27 analogs. Among the various structural modifications, we identified four additional active analogs with enhanced ROCK/MRCK inhibitory potency. The anti-proliferative and cell cycle inhibitory effects of the active analogs were examined in non-small cell lung cancer, breast cancer, and melanoma cell lines. The anti-proliferative effectiveness of DJ4 and the active analogs was further demonstrated against a wide array of cancer cell types using the NCI-60 human cancer cell line panel. Lastly, these new analogs were tested for anti-migratory effects in highly invasive MDA-MB-231 breast cancer cells. Together, our results demonstrate that selective inhibitors of ROCK1/2 (DJE4, DJ-Allyl) inhibited cell proliferation and induced cell cycle arrest at G2/M but were less effective in cell death induction compared with dual ROCK1/2 and MRCKα/β (DJ4 and DJ110).
Collapse
Affiliation(s)
- Vijay Pralhad Kale
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Jeremy A. Hengst
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Arati K. Sharma
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Upendarrao Golla
- Department of Medicine, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Sinisa Dovat
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Shantu G. Amin
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Jong K. Yun
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Dhimant H. Desai
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| |
Collapse
|
7
|
Baran B, Kosieradzka K, Skarzynska W, Niewiadomski P. MRCKα/β positively regulates Gli protein activity. Cell Signal 2023; 107:110666. [PMID: 37019250 DOI: 10.1016/j.cellsig.2023.110666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023]
Abstract
Posttranslational modifications (PTMs) are key regulatory events for the majority of signaling pathways. Transcription factors are often phosphorylated on multiple residues, which regulates their trafficking, stability, or transcriptional activity. Gli proteins, transcription factors that respond to the Hedgehog pathway, are regulated by phosphorylation, but the sites and the kinases involved have been only partially described. We identified three novel kinases: MRCKα, MRCKβ, and MAP4K5 which physically interact with Gli proteins and directly phosphorylate Gli2 on multiple sites. We established that MRCKα/β kinases regulate Gli proteins, which impacts the transcriptional output of the Hedgehog pathway. We showed that double knockout of MRCKα/β affects Gli2 ciliary and nuclear localization and reduces Gli2 binding to the Gli1 promoter. Our research fills a critical gap in our understanding of the regulation of Gli proteins by describing their activation mechanisms through phosphorylation.
Collapse
|
8
|
Santos JC, Profitós-Pelejà N, Sánchez-Vinces S, Roué G. RHOA Therapeutic Targeting in Hematological Cancers. Cells 2023; 12:cells12030433. [PMID: 36766776 PMCID: PMC9914237 DOI: 10.3390/cells12030433] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Primarily identified as an important regulator of cytoskeletal dynamics, the small GTPase Ras homolog gene family member A (RHOA) has been implicated in the transduction of signals regulating a broad range of cellular functions such as cell survival, migration, adhesion and proliferation. Deregulated activity of RHOA has been linked to the growth, progression and metastasis of various cancer types. Recent cancer genome-wide sequencing studies have unveiled both RHOA gain and loss-of-function mutations in primary leukemia/lymphoma, suggesting that this GTPase may exert tumor-promoting or tumor-suppressive functions depending on the cellular context. Based on these observations, RHOA signaling represents an attractive therapeutic target for the development of selective anticancer strategies. In this review, we will summarize the molecular mechanisms underlying RHOA GTPase functions in immune regulation and in the development of hematological neoplasms and will discuss the current strategies aimed at modulating RHOA functions in these diseases.
Collapse
Affiliation(s)
- Juliana Carvalho Santos
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Núria Profitós-Pelejà
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Salvador Sánchez-Vinces
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University Medical School, Braganca Paulista 01246-100, São Paulo, Brazil
| | - Gaël Roué
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
- Correspondence: ; Tel.: +34-935572835
| |
Collapse
|
9
|
Cytoplasmic Clusterin Suppresses Lung Cancer Metastasis by Inhibiting the ROCK1-ERK Axis. Cancers (Basel) 2022; 14:cancers14102463. [PMID: 35626071 PMCID: PMC9140019 DOI: 10.3390/cancers14102463] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary We show that CLU, especially cytoplasmic precursor CLU, is downregulated in lung cancer and correlates with poor survival. The silencing of CLU promotes lung cancer cell migration and invasion, while the overexpression of CLU potently inhibits these phenomena. Interestingly, secretory CLU proteins are slightly decreased in lung cancer tissue and fail to exert similar anti-metastatic effects like cytoplasmic precursor CLU, demonstrating that cytoplasmic precursor CLU is the primary functional isoform of CLU, which exerts the anti-metastatic effects of lung cancer. Mechanistically, cytoplasmic precursor CLU binds ROCK1 to decrease phosphorylation of ERK1/2 by inhibiting the kinase activity of ROCK1, leading to an anti-metastatic effect in lung cancer cells. These findings reveal a novel insight into the function and regulation of cytoplasmic CLU in lung cancer, which might be a potential target for the diagnosis and treatment of metastatic lung cancer. Abstract Clusterin (CLU) is a heterodimeric glycoprotein that has been detected in diverse human tissues and implicated in many cellular processes. Accumulating evidence indicates that the expression of secreted CLU correlates with the progression of cancers. However, the molecular mechanisms underlying its tumor-suppressive roles are incompletely uncovered. In this study, we demonstrate that precursor CLU is widely downregulated in lung cancer tissue, in which secretory CLU proteins are slightly decreased. Impressively, overexpressing CLU potently inhibits the migration, invasion and metastasis of lung cancer cells, whereas silencing CLU promotes this behavior; however, it appears that secretory CLU fails to exert similar anti-metastatic effects. Interestingly, the cytoplasmic precursor CLU binds ROCK1 to abrogate the interaction between ROCK1 and ERK and impair ERK activity, leading to the suppression of lung cancer invasiveness. Meanwhile, the expression of CLU was remarkably diminished in lung cancer bone metastasis loci when compared with subcutaneous tumors in the mouse model and hardly detected in the bone metastasis loci of lung cancer patients when compared with the primary. These findings reveal a novel insight into the function and regulation of cytoplasmic CLU in lung cancer, which might be a potential target for the diagnosis and treatment of metastatic lung cancer.
Collapse
|
10
|
Wu Y, Liu B, Lin W, Zhao R, Han W, Xie J. AAMP promotes colorectal cancer metastasis by suppressing SMURF2-mediated ubiquitination and degradation of RhoA. Mol Ther Oncolytics 2021; 23:515-530. [PMID: 34901393 PMCID: PMC8633529 DOI: 10.1016/j.omto.2021.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 10/27/2021] [Accepted: 11/08/2021] [Indexed: 11/16/2022] Open
Abstract
Metastasis is considered the leading cause of cancer death due to the limited possibilities to therapeutically target this process. How the ubiquitination machinery contributes to metastasis remains underexplored. Angio-associated migratory cell protein (AAMP), a ubiquitously expressed protein involved in cell migration, has been reported to play oncogenic roles in breast and non-small cell lung cancer (NSCLC). However, the role of AAMP in colorectal cancer (CRC) has not been demonstrated. Here, we report that AAMP is aberrantly upregulated in metastatic CRC and that AAMP upregulation is correlated with the poor survival of CRC patients. AAMP knockdown significantly attenuated the migration and invasion of CRC cells, while AAMP overexpression led to the opposite effects. Mechanistically, we identified Ras homolog family member A (RhoA) as a target of AAMP. Smad ubiquitin regulatory factor (SMURF) 2 was previously found to be a CRC suppressor. Notably, we discovered here that SMURF2 acted as an E3 ubiquitin ligase to mediate the ubiquitination and degradation of RhoA. AAMP stabilized RhoA by binding to it and suppressing its SMURF2-mediated ubiquitination and degradation. Subsequently, the level of active RhoA was increased, thereby accelerating CRC cell migration and invasion. These findings indicate a new potential antitumor target for CRC.
Collapse
Affiliation(s)
- Yuhui Wu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Bofang Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Weiqiang Lin
- Laboratory of Cancer Biology, Institute of Clinical Science, The Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Yiwu, Zhejiang 322000, China
| | - Rongjie Zhao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Corresponding author: Weidong Han, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, Zhejiang 310016, China.
| | - Jiansheng Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Corresponding author: Jiansheng Xie, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, Zhejiang 310016, China.
| |
Collapse
|
11
|
Roberts AB, Zhang J, Raj Singh V, Nikolić M, Moeendarbary E, Kamm RD, So PTC, Scarcelli G. Tumor cell nuclei soften during transendothelial migration. J Biomech 2021; 121:110400. [PMID: 33882444 PMCID: PMC8274349 DOI: 10.1016/j.jbiomech.2021.110400] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 02/08/2023]
Abstract
During cancer metastasis, tumor cells undergo significant deformation in order to traverse through endothelial cell junctions in the walls of blood vessels. As cells pass through narrow gaps, smaller than the nuclear diameter, the spatial configuration of chromatin must change along with the distribution of nuclear enzymes. Nuclear stiffness is an important determinant of the ability of cells to undergo transendothelial migration, yet no studies have been conducted to assess whether tumor cell cytoskeletal or nuclear stiffness changes during this critical process in order to facilitate passage. To address this question, we employed two non-contact methods, Brillouin confocal microscopy (BCM) and confocal reflectance quantitative phase microscopy (QPM), to track the changes in mechanical properties of live, transmigrating tumor cells in an in vitro collagen gel platform. Using these two imaging modalities to study transmigrating MDA-MB-231, A549, and A375 cells, we found that both the cells and their nuclei soften upon extravasation and that the nuclear membranes remain soft for at least 24 h. These new data suggest that tumor cells adjust their mechanical properties in order to facilitate extravasation.
Collapse
Affiliation(s)
- Anya B Roberts
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
| | - Jitao Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park 20742, MD, USA
| | - Vijay Raj Singh
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge 02139, MA, USA; Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
| | - Miloš Nikolić
- Maryland Biophysics Program, University of Maryland, College Park, MD 20742, USA
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge 02139, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge 02139, MA, USA.
| | - Peter T C So
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge 02139, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge 02139, MA, USA; Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge 02139, MA, USA.
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, College Park 20742, MD, USA; Maryland Biophysics Program, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
12
|
Abstract
The epithelium forms a smart barrier to the external environment that can remodel whilst maintaining tissue integrity, a feature important for development, homeostasis, and function. Its dysregulation can lead to diseases ranging from cancer to vision loss. Epithelial remodeling requires reorganization of a thin sheet of actomyosin cortex under the plasma membrane of polarized cells that form basolateral contacts with neighboring cells and the extracellular matrix (ECM). Rho GTPases act as spatiotemporal molecular switches in this process, controlling localized actomyosin dynamics. However, the molecular mechanisms that control actomyosin dynamics at the apical cortex are poorly understood. This review focusses on a growing body of evidence that suggest myotonic dystrophy kinase-related Cdc42-binding kinase (MRCK) plays a conserved role in morphogenetic signaling at the apical cortex in diverse cell and tissue remodeling processes. The possible molecular and mechanistic basis for the diverse functions of MRCK at the apical pole will also be discussed.
Collapse
Affiliation(s)
- Ceniz Zihni
- UCL Institute of Ophthalmology, Department of Cell Biology, University College London, London, UK
| |
Collapse
|
13
|
Kwa MQ, Brandao R, Phung TH, Ge J, Scieri G, Brakebusch C. MRCKα Is Dispensable for Breast Cancer Development in the MMTV-PyMT Model. Cells 2021; 10:cells10040942. [PMID: 33921698 PMCID: PMC8073694 DOI: 10.3390/cells10040942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/07/2021] [Accepted: 04/15/2021] [Indexed: 11/16/2022] Open
Abstract
MRCKα is a ubiquitously expressed serine/threonine kinase involved in cell contraction and F-actin turnover, which is highly amplified in human breast cancer and part of a gene expression signature for bad prognosis. Nothing is known about the in vivo function of MRCKα. To explore MRCKα function in development and in breast cancer, we generated mice lacking a functional MRCKα gene. Mice were born close to the Mendelian ratio and showed no obvious phenotype including a normal mammary gland formation. Assessing breast cancer development using the transgenic MMTV-PyMT mouse model, loss of MRCKα did not affect tumor onset, tumor growth and metastasis formation. Deleting MRCKα and its related family member MRCKβ in two triple-negative breast cancer cell lines resulted in reduced invasion of MDA-MB-231 cells, but did not affect migration of 4T1 cells. Further genomic analysis of human breast cancers revealed that MRCKα is frequently co-amplified with the oncogenes ARID4B and AKT3 which might contribute to the prognostic value of MRCKα expression. Collectively, these data suggest that MRCKα might be a prognostic marker for breast cancer, but probably of limited functional importance.
Collapse
MESH Headings
- Actin Depolymerizing Factors/metabolism
- Actins/metabolism
- Animals
- Antigens, Neoplasm/metabolism
- Antigens, Polyomavirus Transforming/metabolism
- Base Sequence
- Carcinogenesis/drug effects
- Carcinogenesis/metabolism
- Carcinogenesis/pathology
- Cell Line, Tumor
- Cell Survival/drug effects
- Collagen/pharmacology
- Disease Models, Animal
- Female
- Gels/pharmacology
- Humans
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Tumor Virus, Mouse/drug effects
- Mammary Tumor Virus, Mouse/physiology
- Mice
- Mice, Knockout
- Mutation/genetics
- Myosins/metabolism
- Myotonin-Protein Kinase/metabolism
- Neoplasm Invasiveness
- Neoplasm Metastasis
- Neoplasm Proteins/metabolism
- Phenotype
- Phosphorylation/drug effects
- Polymerization/drug effects
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Triple Negative Breast Neoplasms/pathology
Collapse
Affiliation(s)
- Mei Qi Kwa
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen, Denmark; (M.Q.K.); (R.B.); (T.H.P.); (J.G.); (G.S.)
| | - Rafael Brandao
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen, Denmark; (M.Q.K.); (R.B.); (T.H.P.); (J.G.); (G.S.)
| | - Trong H. Phung
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen, Denmark; (M.Q.K.); (R.B.); (T.H.P.); (J.G.); (G.S.)
- Centre College, 600 W Walnut St, Danville, KY 40422, USA
| | - Jianfeng Ge
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen, Denmark; (M.Q.K.); (R.B.); (T.H.P.); (J.G.); (G.S.)
- Medical Research Centre (MRC) Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, P.O. Box 197, Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Giuseppe Scieri
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen, Denmark; (M.Q.K.); (R.B.); (T.H.P.); (J.G.); (G.S.)
| | - Cord Brakebusch
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen, Denmark; (M.Q.K.); (R.B.); (T.H.P.); (J.G.); (G.S.)
- Correspondence:
| |
Collapse
|
14
|
Targeting the cytoskeleton against metastatic dissemination. Cancer Metastasis Rev 2021; 40:89-140. [PMID: 33471283 DOI: 10.1007/s10555-020-09936-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023]
Abstract
Cancer is a pathology characterized by a loss or a perturbation of a number of typical features of normal cell behaviour. Indeed, the acquisition of an inappropriate migratory and invasive phenotype has been reported to be one of the hallmarks of cancer. The cytoskeleton is a complex dynamic network of highly ordered interlinking filaments playing a key role in the control of fundamental cellular processes, like cell shape maintenance, motility, division and intracellular transport. Moreover, deregulation of this complex machinery contributes to cancer progression and malignancy, enabling cells to acquire an invasive and metastatic phenotype. Metastasis accounts for 90% of death from patients affected by solid tumours, while an efficient prevention and suppression of metastatic disease still remains elusive. This results in the lack of effective therapeutic options currently available for patients with advanced disease. In this context, the cytoskeleton with its regulatory and structural proteins emerges as a novel and highly effective target to be exploited for a substantial therapeutic effort toward the development of specific anti-metastatic drugs. Here we provide an overview of the role of cytoskeleton components and interacting proteins in cancer metastasis with a special focus on small molecule compounds interfering with the actin cytoskeleton organization and function. The emerging involvement of microtubules and intermediate filaments in cancer metastasis is also reviewed.
Collapse
|
15
|
Shao Y, Tong Z, Wei J, Yang T. LncRNA-zinc finger protein 281 downregulates rho-associated coiled-coil containing protein kinase 1 by upregulating miR-144 in osteosarcoma. Oncol Lett 2020; 20:79. [PMID: 32863912 PMCID: PMC7436886 DOI: 10.3892/ol.2020.11940] [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: 05/27/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
Zinc finger protein 281 (ZNF281) has been characterized as a tumor suppressive lncRNA in glioma. The present study aimed to analyze the functionality of ZNF281 in osteosarcoma (OS). It was demonstrated that ZNF281 was downregulated in OS tissue specimens and predicted the survival of patients with OS. In tissues from patients with OS, ZNF281 was negatively associated with rho-associated coiled-coil containing protein kinase 1 (ROCK1), but positively associated with miR-144. In the U2OS cell line, ZNF281 overexpression mediated the upregulation of miR-44 and downregulation of ROCK1. miR-144 overexpression led to the downregulation of ROCK1, but failed to affect ZNF281. Expression of ZNF281 and miR-144 resulted in decreased cell migration and invasion, while ROCK1 overexpression resulted in increased invasion and migration of OS cells. In addition, ROCK1 overexpression attenuated the effects of ZNF281 and miR-144 overexpression. Thus, ZNF281 may downregulate ROCK1 by upregulating miR-144 and inhibit cancer cell invasion and migration in OS.
Collapse
Affiliation(s)
- Yuxiong Shao
- Department of Bone Disease and Oncology, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Zhichao Tong
- Department of Bone Disease and Oncology, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Jianfeng Wei
- Department of Bone Disease and Oncology, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Tuanmin Yang
- Department of Bone Disease and Oncology, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
16
|
de Sousa GR, Vieira GM, das Chagas PF, Pezuk JA, Brassesco MS. Should we keep rocking? Portraits from targeting Rho kinases in cancer. Pharmacol Res 2020; 160:105093. [PMID: 32726671 DOI: 10.1016/j.phrs.2020.105093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 12/12/2022]
Abstract
Cancer targeted therapy, either alone or in combination with conventional chemotherapy, could allow the survival of patients with neoplasms currently considered incurable. In recent years, the dysregulation of the Rho-associated coiled-coil kinases (ROCK1 and ROCK2) has been associated with increased metastasis and poorer patient survival in several tumor types, and due to their essential roles in regulating the cytoskeleton, have gained popularity and progressively been researched as targets for the development of novel anti-cancer drugs. Nevertheless, in a pediatric scenario, the influence of both isoforms on prognosis remains a controversial issue. In this review, we summarize the functions of ROCKs, compile their roles in human cancer and their value as prognostic factors in both, adult and pediatric cancer. Moreover, we provide the up-to-date advances on their pharmacological inhibition in pre-clinical models and clinical trials. Alternatively, we highlight and discuss detrimental effects of ROCK inhibition provoked not only by the action on off-targets, but most importantly, by pro-survival effects on cancer stem cells, dormant cells, and circulating tumor cells, along with cell-context or microenvironment-dependent contradictory responses. Together these drawbacks represent a risk for cancer cell dissemination and metastasis after anti-ROCK intervention, a caveat that should concern scientists and clinicians.
Collapse
Affiliation(s)
| | | | | | | | - María Sol Brassesco
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Brazil.
| |
Collapse
|
17
|
Dysregulation of Rho GTPases in Human Cancers. Cancers (Basel) 2020; 12:cancers12051179. [PMID: 32392742 PMCID: PMC7281333 DOI: 10.3390/cancers12051179] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 01/28/2023] Open
Abstract
Rho GTPases play central roles in numerous cellular processes, including cell motility, cell polarity, and cell cycle progression, by regulating actin cytoskeletal dynamics and cell adhesion. Dysregulation of Rho GTPase signaling is observed in a broad range of human cancers, and is associated with cancer development and malignant phenotypes, including metastasis and chemoresistance. Rho GTPase activity is precisely controlled by guanine nucleotide exchange factors, GTPase-activating proteins, and guanine nucleotide dissociation inhibitors. Recent evidence demonstrates that it is also regulated by post-translational modifications, such as phosphorylation, ubiquitination, and sumoylation. Here, we review the current knowledge on the role of Rho GTPases, and the precise mechanisms controlling their activity in the regulation of cancer progression. In addition, we discuss targeting strategies for the development of new drugs to improve cancer therapy.
Collapse
|
18
|
Clayton NS, Ridley AJ. Targeting Rho GTPase Signaling Networks in Cancer. Front Cell Dev Biol 2020; 8:222. [PMID: 32309283 PMCID: PMC7145979 DOI: 10.3389/fcell.2020.00222] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/16/2020] [Indexed: 12/16/2022] Open
Abstract
As key regulators of cytoskeletal dynamics, Rho GTPases coordinate a wide range of cellular processes, including cell polarity, cell migration, and cell cycle progression. The adoption of a pro-migratory phenotype enables cancer cells to invade the stroma surrounding the primary tumor and move toward and enter blood or lymphatic vessels. Targeting these early events could reduce the progression to metastatic disease, the leading cause of cancer-related deaths. Rho GTPases play a key role in the formation of dynamic actin-rich membrane protrusions and the turnover of cell-cell and cell-extracellular matrix adhesions required for efficient cancer cell invasion. Here, we discuss the roles of Rho GTPases in cancer, their validation as therapeutic targets and the challenges of developing clinically viable Rho GTPase inhibitors. We review other therapeutic targets in the wider Rho GTPase signaling network and focus on the four best characterized effector families: p21-activated kinases (PAKs), Rho-associated protein kinases (ROCKs), atypical protein kinase Cs (aPKCs), and myotonic dystrophy kinase-related Cdc42-binding kinases (MRCKs).
Collapse
Affiliation(s)
- Natasha S Clayton
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Anne J Ridley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
19
|
Kurimchak AM, Herrera-Montávez C, Brown J, Johnson KJ, Sodi V, Srivastava N, Kumar V, Deihimi S, O'Brien S, Peri S, Mantia-Smaldone GM, Jain A, Winters RM, Cai KQ, Chernoff J, Connolly DC, Duncan JS. Functional proteomics interrogation of the kinome identifies MRCKA as a therapeutic target in high-grade serous ovarian carcinoma. Sci Signal 2020; 13:13/619/eaax8238. [PMID: 32071169 DOI: 10.1126/scisignal.aax8238] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most lethal gynecological cancer with few effective, targeted therapies. HGSOC tumors exhibit genomic instability with frequent alterations in the protein kinome; however, only a small fraction of the kinome has been therapeutically targeted in HGSOC. Using multiplexed inhibitor beads and mass spectrometry, we mapped the kinome landscape of HGSOC tumors from patients and patient-derived xenograft models. The data revealed a prevalent signature consisting of established HGSOC driver kinases, as well as several kinases previously unexplored in HGSOC. Loss-of-function analysis of these kinases in HGSOC cells indicated MRCKA (also known as CDC42BPA) as a putative therapeutic target. Characterization of the effects of MRCKA knockdown in established HGSOC cell lines demonstrated that MRCKA was integral to signaling that regulated the cell cycle checkpoint, focal adhesion, and actin remodeling, as well as cell migration, proliferation, and survival. Moreover, inhibition of MRCKA using the small-molecule BDP9066 decreased cell proliferation and spheroid formation and induced apoptosis in HGSOC cells, suggesting that MRCKA may be a promising therapeutic target for the treatment of HGSOC.
Collapse
Affiliation(s)
- Alison M Kurimchak
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | - Jennifer Brown
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Katherine J Johnson
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Thermo Fisher Scientific, 168 Third Ave., Waltham, MA 02451, USA
| | - Valerie Sodi
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Nishi Srivastava
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Vikas Kumar
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Safoora Deihimi
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Shane O'Brien
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Suraj Peri
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, PA. 19111, USA
| | - Gina M Mantia-Smaldone
- Division of Gynecologic Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Angela Jain
- Division of Gynecologic Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Ryan M Winters
- Biosample Repository Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Denise C Connolly
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - James S Duncan
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
20
|
Villalobo A, Berchtold MW. The Role of Calmodulin in Tumor Cell Migration, Invasiveness, and Metastasis. Int J Mol Sci 2020; 21:ijms21030765. [PMID: 31991573 PMCID: PMC7037201 DOI: 10.3390/ijms21030765] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/18/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Calmodulin (CaM) is the principal Ca2+ sensor protein in all eukaryotic cells, that upon binding to target proteins transduces signals encoded by global or subcellular-specific changes of Ca2+ concentration within the cell. The Ca2+/CaM complex as well as Ca2+-free CaM modulate the activity of a vast number of enzymes, channels, signaling, adaptor and structural proteins, and hence the functionality of implicated signaling pathways, which control multiple cellular functions. A basic and important cellular function controlled by CaM in various ways is cell motility. Here we discuss the role of CaM-dependent systems involved in cell migration, tumor cell invasiveness, and metastasis development. Emphasis is given to phosphorylation/dephosphorylation events catalyzed by myosin light-chain kinase, CaM-dependent kinase-II, as well as other CaM-dependent kinases, and the CaM-dependent phosphatase calcineurin. In addition, the role of the CaM-regulated small GTPases Rac1 and Cdc42 (cell division cycle protein 42) as well as CaM-binding adaptor/scaffold proteins such as Grb7 (growth factor receptor bound protein 7), IQGAP (IQ motif containing GTPase activating protein) and AKAP12 (A kinase anchoring protein 12) will be reviewed. CaM-regulated mechanisms in cancer cells responsible for their greater migratory capacity compared to non-malignant cells, invasion of adjacent normal tissues and their systemic dissemination will be discussed, including closely linked processes such as the epithelial–mesenchymal transition and the activation of metalloproteases. This review covers as well the role of CaM in establishing metastatic foci in distant organs. Finally, the use of CaM antagonists and other blocking techniques to downregulate CaM-dependent systems aimed at preventing cancer cell invasiveness and metastasis development will be outlined.
Collapse
Affiliation(s)
- Antonio Villalobo
- Cancer and Human Molecular Genetics Area—Oto-Neurosurgery Research Group, University Hospital La Paz Research Institute (IdiPAZ), Paseo de la Castellana 261, E-28046 Madrid, Spain
- Correspondence: (A.V.); (M.W.B.)
| | - Martin W. Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen, Denmark
- Correspondence: (A.V.); (M.W.B.)
| |
Collapse
|
21
|
Identification of Kinases Responsible for p53-Dependent Autophagy. iScience 2019; 15:109-118. [PMID: 31048145 PMCID: PMC6495467 DOI: 10.1016/j.isci.2019.04.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/11/2019] [Accepted: 04/17/2019] [Indexed: 01/03/2023] Open
Abstract
In cancer, autophagy is upregulated to promote cell survival and tumor growth during times of nutrient stress and can confer resistance to drug treatments. Several major signaling networks control autophagy induction, including the p53 tumor suppressor pathway. In response to DNA damage and other cellular stresses, p53 is stabilized and activated, while HDM2 binds to and ubiquitinates p53 for proteasome degradation. Thus blocking the HDM2-p53 interaction is a promising therapeutic strategy in cancer; however, the potential survival advantage conferred by autophagy induction may limit therapeutic efficacy. In this study, we leveraged an HDM2 inhibitor to identify kinases required for p53-dependent autophagy. Interestingly, we discovered that p53-dependent autophagy requires several kinases, including the myotonic dystrophy protein kinase-like alpha (MRCKα). MRCKα is a CDC42 effector reported to activate actin-myosin cytoskeletal reorganization. Overall, this study provides evidence linking MRCKα to autophagy and reveals additional insights into the role of kinases in p53-dependent autophagy. HDM2 inhibitors stabilize and activate p53 leading to robust autophagy induction RNAi screen uncovers kinases involved in p53-dependent autophagy ULK1 and the actin cytoskeleton kinase MRCKα mediate p53-induced autophagy
Collapse
|
22
|
Eble JA, Niland S. The extracellular matrix in tumor progression and metastasis. Clin Exp Metastasis 2019; 36:171-198. [PMID: 30972526 DOI: 10.1007/s10585-019-09966-1] [Citation(s) in RCA: 341] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) constitutes the scaffold of tissues and organs. It is a complex network of extracellular proteins, proteoglycans and glycoproteins, which form supramolecular aggregates, such as fibrils and sheet-like networks. In addition to its biochemical composition, including the covalent intermolecular cross-linkages, the ECM is also characterized by its biophysical parameters, such as topography, molecular density, stiffness/rigidity and tension. Taking these biochemical and biophysical parameters into consideration, the ECM is very versatile and undergoes constant remodeling. This review focusses on this remodeling of the ECM under the influence of a primary solid tumor mass. Within this tumor stroma, not only the cancer cells but also the resident fibroblasts, which differentiate into cancer-associated fibroblasts (CAFs), modify the ECM. Growth factors and chemokines, which are tethered to and released from the ECM, as well as metabolic changes of the cells within the tumor bulk, add to the tumor-supporting tumor microenvironment. Metastasizing cancer cells from a primary tumor mass infiltrate into the ECM, which variably may facilitate cancer cell migration or act as barrier, which has to be proteolytically breached by the infiltrating tumor cell. The biochemical and biophysical properties therefore determine the rates and routes of metastatic dissemination. Moreover, primed by soluble factors of the primary tumor, the ECM of distant organs may be remodeled in a way to facilitate the engraftment of metastasizing cancer cells. Such premetastatic niches are responsible for the organotropic preference of certain cancer entities to colonize at certain sites in distant organs and to establish a metastasis. Translational application of our knowledge about the cancer-primed ECM is sparse with respect to therapeutic approaches, whereas tumor-induced ECM alterations such as increased tissue stiffness and desmoplasia, as well as breaching the basement membrane are hallmark of malignancy and diagnostically and histologically harnessed.
Collapse
Affiliation(s)
- Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany.
| | - Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| |
Collapse
|
23
|
Unbekandt M, Belshaw S, Bower J, Clarke M, Cordes J, Crighton D, Croft DR, Drysdale MJ, Garnett MJ, Gill K, Gray C, Greenhalgh DA, Hall JAM, Konczal J, Lilla S, McArthur D, McConnell P, McDonald L, McGarry L, McKinnon H, McMenemy C, Mezna M, Morrice NA, Munro J, Naylor G, Rath N, Schüttelkopf AW, Sime M, Olson MF. Discovery of Potent and Selective MRCK Inhibitors with Therapeutic Effect on Skin Cancer. Cancer Res 2018; 78:2096-2114. [PMID: 29382705 PMCID: PMC5901721 DOI: 10.1158/0008-5472.can-17-2870] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/20/2017] [Accepted: 01/24/2018] [Indexed: 01/29/2023]
Abstract
The myotonic dystrophy-related Cdc42-binding kinases MRCKα and MRCKβ contribute to the regulation of actin-myosin cytoskeleton organization and dynamics, acting in concert with the Rho-associated coiled-coil kinases ROCK1 and ROCK2. The absence of highly potent and selective MRCK inhibitors has resulted in relatively little knowledge of the potential roles of these kinases in cancer. Here, we report the discovery of the azaindole compounds BDP8900 and BDP9066 as potent and selective MRCK inhibitors that reduce substrate phosphorylation, leading to morphologic changes in cancer cells along with inhibition of their motility and invasive character. In over 750 human cancer cell lines tested, BDP8900 and BDP9066 displayed consistent antiproliferative effects with greatest activity in hematologic cancer cells. Mass spectrometry identified MRCKα S1003 as an autophosphorylation site, enabling development of a phosphorylation-sensitive antibody tool to report on MRCKα status in tumor specimens. In a two-stage chemical carcinogenesis model of murine squamous cell carcinoma, topical treatments reduced MRCKα S1003 autophosphorylation and skin papilloma outgrowth. In parallel work, we validated a phospho-selective antibody with the capability to monitor drug pharmacodynamics. Taken together, our findings establish an important oncogenic role for MRCK in cancer, and they offer an initial preclinical proof of concept for MRCK inhibition as a valid therapeutic strategy.Significance: The development of selective small-molecule inhibitors of the Cdc42-binding MRCK kinases reveals their essential roles in cancer cell viability, migration, and invasive character. Cancer Res; 78(8); 2096-114. ©2018 AACR.
Collapse
Affiliation(s)
- Mathieu Unbekandt
- Molecular Cell Biology Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Simone Belshaw
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Justin Bower
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Maeve Clarke
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Jacqueline Cordes
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Diane Crighton
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Daniel R Croft
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Martin J Drysdale
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Mathew J Garnett
- Translational Cancer Genomics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Kathryn Gill
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Christopher Gray
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - David A Greenhalgh
- Section of Dermatology and Molecular Carcinogenesis, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - James A M Hall
- Translational Cancer Genomics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Jennifer Konczal
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Sergio Lilla
- Mass Spectrometry Facility, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Duncan McArthur
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Patricia McConnell
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Laura McDonald
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Lynn McGarry
- Screening Facility, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Heather McKinnon
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Carol McMenemy
- Section of Dermatology and Molecular Carcinogenesis, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mokdad Mezna
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Nicolas A Morrice
- Mass Spectrometry Facility, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - June Munro
- Molecular Cell Biology Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Gregory Naylor
- Molecular Cell Biology Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Nicola Rath
- Molecular Cell Biology Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | | | - Mairi Sime
- Drug Discovery Unit, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Michael F Olson
- Molecular Cell Biology Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom.
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
24
|
Abstract
Malignant carcinomas are often characterized by metastasis, the movement of carcinoma cells from a primary site to colonize distant organs. For metastasis to occur, carcinoma cells first must adopt a pro-migratory phenotype and move through the surrounding stroma towards a blood or lymphatic vessel. Currently, there are very limited possibilities to target these processes therapeutically. The family of Rho GTPases is an ubiquitously expressed division of GTP-binding proteins involved in the regulation of cytoskeletal dynamics and intracellular signaling. The best characterized members of the Rho family GTPases are RhoA, Rac1 and Cdc42. Abnormalities in Rho GTPase function have major consequences for cancer progression. Rho GTPase activation is driven by cell surface receptors that activate GTP exchange factors (GEFs) and GTPase-activating proteins (GAPs). In this review, we summarize our current knowledge on Rho GTPase function in the regulation of metastasis. We will focus on key discoveries in the regulation of epithelial-mesenchymal-transition (EMT), cell-cell junctions, formation of membrane protrusions, plasticity of cell migration and adaptation to a hypoxic environment. In addition, we will emphasize on crosstalk between Rho GTPase family members and other important oncogenic pathways, such as cyclic AMP-mediated signaling, canonical Wnt/β-catenin, Yes-associated protein (YAP) and hypoxia inducible factor 1α (Hif1α) and provide an overview of the advancements and challenges in developing pharmacological tools to target Rho GTPase and the aforementioned crosstalk in the context of cancer therapeutics.
Collapse
|
25
|
Cardama GA, Gonzalez N, Maggio J, Menna PL, Gomez DE. Rho GTPases as therapeutic targets in cancer (Review). Int J Oncol 2017; 51:1025-1034. [PMID: 28848995 PMCID: PMC5592879 DOI: 10.3892/ijo.2017.4093] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022] Open
Abstract
Rho GTPases are key molecular switches controlling the transduction of external signals to cytoplasmic and nuclear effectors. In the last few years, the development of genetic and pharmacological tools has allowed a more precise definition of the specific roles of Rho GTPases in cancer. The aim of the present review is to describe the cellular functions regulated by these proteins with focus in deregulated signals present in malignant tumors. Finally, we describe the state of the art in search of different experimental therapeutic strategies with Rho GTPases as molecular targets.
Collapse
Affiliation(s)
- G A Cardama
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - N Gonzalez
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - J Maggio
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - P Lorenzano Menna
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - D E Gomez
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| |
Collapse
|
26
|
Functions of Rho family of small GTPases and Rho-associated coiled-coil kinases in bone cells during differentiation and mineralization. Biochim Biophys Acta Gen Subj 2017; 1861:1009-1023. [PMID: 28188861 DOI: 10.1016/j.bbagen.2017.02.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 02/02/2017] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Members of Rho-associated coiled-coil kinases (ROCKs) are effectors of Rho family of small GTPases. ROCKs have multiple functions that include regulation of cellular contraction and polarity, adhesion, motility, proliferation, apoptosis, differentiation, maturation and remodeling of the extracellular matrix (ECM). SCOPE OF THE REVIEW Here, we focus on the action of RhoA and RhoA effectors, ROCK1 and ROCK2, in cells related to tissue mineralization: mesenchymal stem cells, chondrocytes, preosteoblasts, osteoblasts, osteocytes, lining cells and osteoclasts. MAJOR CONCLUSIONS The activation of the RhoA/ROCK pathway promotes stress fiber formation and reduces chondrocyte and osteogenic differentiations, in contrast to that in mesenchymal stem cells which stimulated the osteogenic and the chondrogenic differentiation. The effects of Rac1 and Cdc42 in promoting chondrocyte hypertrophy and of Rac1, Rac2 and Cdc42 in osteoclast are discussed. In addition, members of the Rho family of GTPases such Rac1, Rac2, Rac3 and Cdc42, acting upstream of ROCK and/or other protein effectors, may compensate the actions of RhoA, affecting directly or indirectly the actions of ROCKs as well as other protein effectors. GENERAL SIGNIFICANCE ROCK activity can trigger cartilage degradation and affect bone formation, therefore these kinases may represent a possible therapeutic target to treat osteoarthritis and osseous diseases. Inhibition of Rho/ROCK activity in chondrocytes prevents cartilage degradation, stimulate mineralization of osteoblasts and facilitate bone formation around implanted metals. Treatment with osteoprotegerin results in a significant decrease in the expression of Rho GTPases, ROCK1 and ROCK2, reducing bone resorption. Inhibition of ROCK signaling increases osteoblast differentiation in a topography-dependent manner.
Collapse
|
27
|
Croze RH, Thi WJ, Clegg DO. ROCK Inhibition Promotes Attachment, Proliferation, and Wound Closure in Human Embryonic Stem Cell-Derived Retinal Pigmented Epithelium. Transl Vis Sci Technol 2016; 5:7. [PMID: 27917311 PMCID: PMC5132148 DOI: 10.1167/tvst.5.6.7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 10/10/2016] [Indexed: 12/27/2022] Open
Abstract
Purpose Nonexudative (dry) age-related macular degeneration (AMD), a leading cause of blindness in the elderly, is associated with the loss of retinal pigmented epithelium (RPE) cells and the development of geographic atrophy, which are areas devoid of RPE cells and photoreceptors. One possible treatment option would be to stimulate RPE attachment and proliferation to replace dying/dysfunctional RPE and bring about wound repair. Clinical trials are underway testing injections of RPE cells derived from pluripotent stem cells to determine their safety and efficacy in treating AMD. However, the factors regulating RPE responses to AMD-associated lesions are not well understood. Here, we use cell culture to investigate the role of RhoA coiled coil kinases (ROCKs) in human embryonic stem cell–derived RPE (hESC-RPE) attachment, proliferation, and wound closure. Methods H9 hESC were spontaneously differentiated into RPE cells. hESC-RPE cells were treated with a pan ROCK1/2 or a ROCK2 only inhibitor; attachment, and proliferation and cell size within an in vitro scratch assay were examined. Results Pharmacological inhibition of ROCKs promoted hESC-RPE attachment and proliferation, and increased the rate of closure of in vitro wounds. ROCK inhibition decreased phosphorylation of cofilin and myosin light chain, suggesting that regulation of the cytoskeleton underlies the mechanism of action of ROCK inhibition. Conclusions ROCK inhibition promotes attachment, proliferation, and wound closure in H9 hESC-RPE cells. ROCK isoforms may have different roles in wound healing. Translational Relevance Modulation of the ROCK-cytoskeletal axis has potential in stimulating wound repair in transplanted RPE cells and attachment in cellular therapies.
Collapse
Affiliation(s)
- Roxanne H Croze
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA, USA
| | - William J Thi
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Dennis O Clegg
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA, USA
| |
Collapse
|
28
|
Lee M, Koh W, Kim B, Chung H, Cho G, Kim H. Involvement of cAMP in the Human Serum-Induced Migration of Adipose-Derived Stem Cells. Dev Reprod 2016; 20:123-30. [PMID: 27660827 PMCID: PMC5027217 DOI: 10.12717/dr.2016.20.2.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Previously we observed that human adipose-derived stem cells (hADSCs) could form aggregation during culture in the presence of human serum (HS). In the present study, we have examined if the aggregation might result from the cell migration and analyzed the difference of cell adhesivity after culture in various conditions. When cells were cultured in fetal bovine serum (FBS) alone, there was no morphological change. Similarly, cells pretreated with FBS for 1 day or cultured in a mixture of FBS and HS showed little change. In contrast, cells cultured in HS alone exhibited formation of cell-free area (spacing) and/or cell aggregation. When cells cultured in FBS or pretreated with FBS were treated with 0.06% trypsin, almost cells remained attached to the dish surfaces. In contrast, when cells cultured in HS alone were examined, most cells detached from the dish by the same treatment. Treatment of cells with forskolin, isobutylmethyl xanthine (IBMX) or LY294002 inhibited the formation of spacing whereas H89 or Y27632 showed little effect. When these cells were treated with 0.06% trypsin after culture, most cells detached from the dishes as cells cultured in HS alone did. However, cells treated with IBMX exhibited weaker adhesivity than HS alone. Based on these observations, it is suggested that HS treatment might decrease the adhesivity and induce three-dimensional migration of hADSCs, in the latter of which cAMP signaling could be involved.
Collapse
Affiliation(s)
- Minji Lee
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - Wonyoung Koh
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - Bomee Kim
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - Hyeju Chung
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - Gahyang Cho
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - Haekwon Kim
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| |
Collapse
|
29
|
Iskit S, Lieftink C, Halonen P, Shahrabi A, Possik PA, Beijersbergen RL, Peeper DS. Integrated in vivo genetic and pharmacologic screening identifies co-inhibition of EGRF and ROCK as a potential treatment regimen for triple-negative breast cancer. Oncotarget 2016; 7:42859-42872. [PMID: 27374095 PMCID: PMC5189992 DOI: 10.18632/oncotarget.10230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is the second most common cause of cancer-related deaths worldwide among women. Despite several therapeutic options, 15% of breast cancer patients succumb to the disease owing to tumor relapse and acquired therapy resistance. Particularly in triple-negative breast cancer (TNBC), developing effective treatments remains challenging owing to the lack of a common vulnerability that can be exploited by targeted approaches. We have previously shown that tumor cells have different requirements for growth in vivo than in vitro. Therefore, to discover novel drug targets for TNBC, we performed parallel in vivo and in vitro genetic shRNA dropout screens. We identified several potential drug targets that were required for tumor growth in vivo to a greater extent than in vitro. By combining pharmacologic inhibitors acting on a subset of these candidates, we identified a synergistic interaction between EGFR and ROCK inhibitors. This combination effectively reduced TNBC cell growth by inducing cell cycle arrest. These results illustrate the power of in vivo genetic screens and warrant further validation of EGFR and ROCK as combined pharmacologic targets for breast cancer.
Collapse
Affiliation(s)
- Sedef Iskit
- Department of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan, Amsterdam, The Netherlands
| | - Cor Lieftink
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan, Amsterdam, The Netherlands
| | - Pasi Halonen
- Drug Discovery Research and Screening Services, BioFocus, Darwinweg, Leiden
| | - Aida Shahrabi
- Department of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan, Amsterdam, The Netherlands
| | | | - Roderick L. Beijersbergen
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan, Amsterdam, The Netherlands
| | - Daniel S. Peeper
- Department of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Abstract
How to design a ligand to bind multiple targets, rather than to a single target, is the focus of this review. Rational polypharmacology draws on knowledge that is both broad ranging and hierarchical. Computer-aided multitarget ligand design methods are described according to their nested knowledge level. Ligand-only and then receptor-ligand strategies are first described; followed by the metabolic network viewpoint. Subsequently strategies that view infectious diseases as multigenomic targets are discussed, and finally the disease level interpretation of medicinal therapy is considered. As yet there is no consensus on how best to proceed in designing a multitarget ligand. The current methodologies are bought together in an attempt to give a practical overview of how polypharmacology design might be best initiated.
Collapse
|
31
|
Wei L, Surma M, Shi S, Lambert-Cheatham N, Shi J. Novel Insights into the Roles of Rho Kinase in Cancer. Arch Immunol Ther Exp (Warsz) 2016; 64:259-78. [PMID: 26725045 PMCID: PMC4930737 DOI: 10.1007/s00005-015-0382-6] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 11/24/2015] [Indexed: 12/12/2022]
Abstract
Rho-associated coiled-coil kinase (ROCK) is a major downstream effector of the small GTPase RhoA. The ROCK family, consisting of ROCK1 and ROCK2, plays a central role in the organization of the actin cytoskeleton, and is involved in a wide range of fundamental cellular functions such as contraction, adhesion, migration, proliferation, and apoptosis. Since the discovery of effective inhibitors such as fasudil and Y27632, the biological roles of ROCK have been extensively explored in numerous diseases, including cancer. Accumulating evidence supports the concept that ROCK plays important roles in tumor development and progression through regulating many key cellular functions associated with malignancy, including tumorigenicity, tumor growth, metastasis, angiogenesis, tumor cell apoptosis/survival and chemoresistance as well. This review focuses on the new advances of the most recent 5 years from the studies on the roles of ROCK in cancer development and progression; the discussion is mainly focused on the potential value of ROCK inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Lei Wei
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA. .,Department of Cellular and Integrative Physiology, Indiana University, School of Medicine, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| | - Michelle Surma
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA
| | - Stephanie Shi
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA
| | - Nathan Lambert-Cheatham
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA
| | - Jianjian Shi
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
32
|
Zucchini C, Martinelli M, De Sanctis P, Rodia MT, Mattei G, Ugolini G, Montroni I, Ghignone F, Solmi R. Possible Gender-Related Modulation by the ROCK1 Gene in Colorectal Cancer Susceptibility. Pathobiology 2015; 82:252-8. [PMID: 26562026 DOI: 10.1159/000439405] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/12/2015] [Indexed: 11/19/2022] Open
Abstract
AIM In view of accumulating evidence supporting a pivotal role of the Rho/ROCK pathway in cancer, we investigated Rho-kinase polymorphisms as potential susceptibility factors in colorectal cancer (CRC) in a representative sample of the Italian population. METHODS DNA obtained from the peripheral blood samples of 137 CRC patients and 141 healthy controls was genotyped for four ROCK1 (rs35996865; rs73963110; rs2127958; rs288980) and five ROCK2 (rs12692437; rs7563468; rs35768389; rs17463896; rs16857265) selected single nucleotide polymorphisms. RESULTS None of the allelic variants of the nine selected markers was associated with the occurrence of CRC or with the development of regional lymph node metastasis. By contrast, the ROCK1 rs35996865 G variant allele was significantly more frequent in male patients (p = 0.028) than in the control group. CONCLUSION This finding is, at present, the first that points to a possible gender-related modulation by the ROCK1 gene in CRC susceptibility.
Collapse
Affiliation(s)
- Cinzia Zucchini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mikami T, Yoshida K, Sawada H, Esaki M, Yasumura K, Ono M. Inhibition of Rho-associated kinases disturbs the collective cell migration of stratified TE-10 cells. Biol Res 2015; 48:48. [PMID: 26330114 PMCID: PMC4556056 DOI: 10.1186/s40659-015-0039-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 08/12/2015] [Indexed: 11/10/2022] Open
Abstract
Background The collective cell migration of stratified epithelial cells is considered to be an important phenomenon in wound healing, development, and cancer invasion; however, little is known about the mechanisms involved. Furthermore, whereas Rho family proteins, including RhoA, play important roles in cell migration, the exact role of Rho-associated coiled coil-containing protein kinases (ROCKs) in cell migration is controversial and might be cell-type dependent. Here, we report the development of a novel modified scratch assay that was used to observe the collective cell migration of stratified TE-10 cells derived from a human esophageal cancer specimen. Results Desmosomes were found between the TE-10 cells and microvilli of the surface of the cell sheet. The leading edge of cells in the cell sheet formed a simple layer and moved forward regularly; these rows were followed by the stratified epithelium. ROCK inhibitors and ROCK small interfering RNAs (siRNAs) disturbed not only the collective migration of the leading edge of this cell sheet, but also the stratified layer in the rear. In contrast, RhoA siRNA treatment resulted in more rapid migration of the leading rows and disturbed movement of the stratified portion. Conclusions The data presented in this study suggest that ROCKs play an important role in mediating the collective migration of TE-10 cell sheets. In addition, differences between the effects of siRNAs targeting either RhoA or ROCKs suggested that distinct mechanisms regulate the collective cell migration in the simple epithelium of the wound edge versus the stratified layer of the epithelium. Electronic supplementary material The online version of this article (doi:10.1186/s40659-015-0039-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Taro Mikami
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, Kanagawa-ken, Japan. .,Department of Plastic and Reconstructive Surgery, Fujisawa Shounandai Hospital, Fujisawa, Kanagawa-ken, Japan. .,Department of Plastic and Reconstructive Surgery, Yokohama City University Hospital, Yokohama, Kanagawa-ken, Japan.
| | - Keiichiro Yoshida
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, Kanagawa-ken, Japan.
| | - Hajime Sawada
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, Kanagawa-ken, Japan.
| | - Michiyo Esaki
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, Kanagawa-ken, Japan.
| | - Kazunori Yasumura
- Department of Plastic and Reconstructive Surgery, Yokohama City University Hospital, Yokohama, Kanagawa-ken, Japan.
| | - Michio Ono
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, Kanagawa-ken, Japan.
| |
Collapse
|