1
|
Ha JH, Radhakrishnan R, Nadhan R, Gomathinayagam R, Jayaraman M, Yan M, Kashyap S, Fung KM, Xu C, Bhattacharya R, Mukherjee P, Isidoro C, Song YS, Dhanasekaran DN. Deciphering a GPCR-lncrna-miRNA nexus: Identification of an aberrant therapeutic target in ovarian cancer. Cancer Lett 2024; 591:216891. [PMID: 38642607 DOI: 10.1016/j.canlet.2024.216891] [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: 02/07/2024] [Revised: 03/30/2024] [Accepted: 04/11/2024] [Indexed: 04/22/2024]
Abstract
Ovarian cancer ranks as a leading cause of mortality among gynecological malignancies, primarily due to the lack of early diagnostic tools, effective targeted therapy, and clear understanding of disease etiology. Previous studies have identified the pivotal role of Lysophosphatidic acid (LPA)-signaling in ovarian cancer pathobiology. Our earlier transcriptomic analysis identified Urothelial Carcinoma Associated-1 (UCA1) as an LPA-stimulated long non-coding RNA (lncRNA). In this study, we elucidate the tripartite interaction between LPA-signaling, UCA1, and let-7 miRNAs in ovarian cancer progression. Results show that the elevated expression of UCA1 enhances cell proliferation, invasive migration, and therapy resistance in high-grade serous ovarian carcinoma cells, whereas silencing UCA1 reverses these oncogenic phenotypes. UCA1 expression inversely correlates with survival outcomes and therapy response in ovarian cancer clinical samples, underscoring its prognostic significance. Mechanistically, UCA1 sequesters let-7 miRNAs, effectively neutralizing their tumor-suppressive functions involving key oncogenes such as Ras and c-Myc. More significantly, intratumoral delivery of UCA1-specific siRNAs inhibits the growth of cisplatin-refractory ovarian cancer xenografts, demonstrating the therapeutic potential of targeting LPAR-UCA1-let-7 axis in ovarian cancer. Thus, our results identify LPAR-UCA1-let-7 axis as a novel avenue for targeted treatment strategies.
Collapse
Affiliation(s)
- Ji Hee Ha
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | | | - Revathy Nadhan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Rohini Gomathinayagam
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Muralidharan Jayaraman
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Mingda Yan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Srishti Kashyap
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Kar-Ming Fung
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Resham Bhattacharya
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Priyabrata Mukherjee
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Ciro Isidoro
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Yong Sang Song
- Seoul National University, College of Medicine, Seoul, 151-921, South Korea
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
2
|
Lee MK, Zhang X, Kim HJ, Hwang YS. Peroxiredoxin 5 is involved in cancer cell invasion and tumor growth of oral squamous cell carcinoma. Oral Dis 2023; 29:423-435. [PMID: 33969595 DOI: 10.1111/odi.13910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/27/2021] [Accepted: 05/02/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Peroxiredoxins (Prxs) are antioxidant enzymes that can coordinate cell signal transduction via reactive species scavenging or by acting as redox sensors. The mechanism by which Prxs promote cancer invasion and progression is not yet fully understood. This study aims to elucidate the precise mechanism through which Prx type 5 (Prx5) promotes cancer invasion and tumor growth. MATERIALS AND METHODS We analyzed the Prx5 expression in oral squamous cell carcinoma (OSCC) by using microarray analysis for gene expression profiling. To identify Prx5 function in cancer, lentiviral short hairpin RNA was used for Prx5 depletion, and invasion assay and mouse xenograft were performed. RESULTS In microarray data obtained from OSCC patients, Prx5 showed higher expression at the tumor margin (TM) compared to the tumor center (TC) of the collective invasion. The depletion of Prx5 in OSCC cells (Prx5dep ) led to decreased invasion activity. In orthotopic xenograft models, Prx5dep cells harbored delimited tumorigenicity compared to wild-type cells as well as the suppression of lymph node metastasis. Prx5dep cells showed growth retardation and increased cellular reactive oxygen species (ROS) levels. The growth retardation of Prx5dep cells resulted in G1 phase arrest. CONCLUSIONS This study provides evidence that Prx5 removes excess ROS, especially in the TM, contributing to cancer invasion and tumor progression.
Collapse
Affiliation(s)
- Min Kyeong Lee
- Department of Dental Hygiene, College of Health Science, Eulji University, Republic of Korea
| | - Xianglan Zhang
- Department of Pathology, Yanbian University Hospital, Yanji, China.,Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Hyung Jun Kim
- Department of Oral Maxillofacial Surgery, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Young Sun Hwang
- Department of Dental Hygiene, College of Health Science, Eulji University, Republic of Korea
| |
Collapse
|
3
|
Lysophosphatidic Acid Signaling in Cancer Cells: What Makes LPA So Special? Cells 2021; 10:cells10082059. [PMID: 34440828 PMCID: PMC8394178 DOI: 10.3390/cells10082059] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/13/2022] Open
Abstract
Lysophosphatidic acid (LPA) refers to a family of simple phospholipids that act as ligands for G protein-coupled receptors. While LPA exerts effects throughout the body in normal physiological circumstances, its pathological role in cancer is of great interest from a therapeutic viewpoint. The numerous LPA receptors (LPARs) are coupled to a variety of G proteins, and more than one LPAR is typically expressed on any given cell. While the individual receptors signal through conventional GPCR pathways, LPA is particularly efficacious in stimulating cancer cell proliferation and migration. This review addresses the mechanistic aspects underlying these pro-tumorigenic effects. We provide examples of LPA signaling responses in various types of cancers, with an emphasis on those where roles have been identified for specific LPARs. While providing an overview of LPAR signaling, these examples also reveal gaps in our knowledge regarding the mechanisms of LPA action at the receptor level. The current understanding of the LPAR structure and the roles of LPAR interactions with other receptors are discussed. Overall, LPARs provide insight into the potential molecular mechanisms that underlie the ability of individual GPCRs (or combinations of GPCRs) to elicit a unique spectrum of responses from their agonist ligands. Further knowledge of these mechanisms will inform drug discovery, since GPCRs are promising therapeutic targets for cancer.
Collapse
|
4
|
Mughees M, Bano F, Wajid S. Mechanism of WASP and WAVE family proteins in the progression of prostate cancer. PROTOPLASMA 2021; 258:683-693. [PMID: 33471226 DOI: 10.1007/s00709-021-01608-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
Prostate cancer (PCa) is the second most commonly diagnosed and third lethal cause of death from cancer in men worldwide. Despite the availability of vast treatment procedures, still the high occurrence of invasion and metastasis of PCa are reported in cancer patients. The WASP (Wiskott-Aldrich syndrome protein) and WAVE (WASP family verprolin homologous protein) family of proteins are actin cytoskeleton regulatory proteins, reported to enhance cancer cell invasion and migration in prostate cancer. Hence, this review sheds light on the studies that explored the potential role of WASP and WAVE family of proteins in invasion and metastasis of prostate cancer. The research articles explored for the completion of this review were mostly from PubMed and Google Scholar by using the appropriate keywords for indexing. The conserved function of WASP and WAVE protein family is to receive the upstream signals from the Rho GTPase family and transmit them to activate the Arp2/3 complex that leads to rapid actin polymerization at leading edge of cells, which is crucial for PCa metastasis. Therefore, targeting these proteins could reflect a very interesting therapeutic opportunity to combat prostate cancer.
Collapse
Affiliation(s)
- Mohd Mughees
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Faizia Bano
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Saima Wajid
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
5
|
ROCK Inhibition as Potential Target for Treatment of Pulmonary Hypertension. Cells 2021; 10:cells10071648. [PMID: 34209333 PMCID: PMC8303917 DOI: 10.3390/cells10071648] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a cardiovascular disease caused by extensive vascular remodeling in the lungs, which ultimately leads to death in consequence of right ventricle (RV) failure. While current drugs for PH therapy address the sustained vasoconstriction, no agent effectively targets vascular cell proliferation and tissue inflammation. Rho-associated protein kinases (ROCKs) emerged in the last few decades as promising targets for PH therapy, since ROCK inhibitors demonstrated significant anti-remodeling and anti-inflammatory effects. In this review, current aspects of ROCK inhibition therapy are discussed in relation to the treatment of PH and RV dysfunction, from cell biology to preclinical and clinical studies.
Collapse
|
6
|
Zhai G, Liang W, Xu Y. High Expression of Lysophosphatidic Acid Induces Nerve Injury in LSS Patients via AKT Mediated NF-κB p65 Pathway. Front Pharmacol 2021; 12:641435. [PMID: 33815123 PMCID: PMC8012901 DOI: 10.3389/fphar.2021.641435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/05/2021] [Indexed: 01/22/2023] Open
Abstract
Lumbar spinal stenosis (LSS) is a spinal degenerative disease, complicated with nerve injury. Lysophosphatidic acid (LPA), a kind of glycerophospholipid molecule is elevated in the initial stages of neural injury. This research aimed to investigate the patho-mechanism of nerve injury caused by LPA in LSS patients. Twenty-five LSS patients and fifteen idiopathic scoliosis patients (without neurological symptoms) were recruited from Xianyang Central Hospital of Shanxi Province. We measured the concentration of LPA in cerebrospinal fluid samples of all subjects. Different concentrations (0.1, 1, and 10 mol/L) of LPA were used to stimulate Rat Neurons-spinal cord (RN-SC) cells. The effects of LPA on cell injury was detected by MTT and LDH (lactate dehydrogenase) assay. Cell apoptosis was determined by FCM (flow cytometry) and TUNEL staining. The changes in the expression of key proteins involved in Akt mediated NF-κB p65 pathway intervened by LPA were determined by western blot. RN-SC cells were pretreated with JSH-23 (NF-κB inhibitor) before LPA exposure, followed by cell apoptosis measurement. The concentration of LPA in LSS patients was notably higher than that in control patients (p < 0.01). The level of LPA was positively correlated with the severity of LSS. LPA treatment induced RN-SC cells displaying oval or rounded cell body with degenerated protrusion dose dependently. In addition, LPA decreased RN-SC cell viability and promoted cell apoptosis in a dose-dependent manner. LPA initiated Akt phosphorylation, IKB phosphorylation, and NF-κB nuclear translocation in a dose-dependent manner. However, JSH-23 (NF-κB inhibitor) pre-treatment prevented effects of LPA. The high levels of LPA induced nerve injury by reducing the viability of RN-SC cells and promoted cell apoptosis through Akt mediated NF-κB p65 signaling pathway. LPA might be a new therapeutic target for relieving nerve injury in LSS patients.
Collapse
Affiliation(s)
- Guiliang Zhai
- Orthopedic Surgery, Binzhou Central Hospital of Shandong Province, Binzhou, China
| | - Wenfei Liang
- Department of Stomatology, Binzhou Central Hospital of Shandong Province, Binzhou, ,China
| | - Yongjun Xu
- Xianyang Central Hospital of Shaanxi Province, Xianyang, China
| |
Collapse
|
7
|
Abdul Rahman M, Tan ML, Johnson SP, Hollows RJ, Chai WL, Mansell JP, Yap LF, Paterson IC. Deregulation of lysophosphatidic acid metabolism in oral cancer promotes cell migration via the up-regulation of COX-2. PeerJ 2020; 8:e10328. [PMID: 33240646 PMCID: PMC7666559 DOI: 10.7717/peerj.10328] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the sixth most common cancer worldwide and accounts for 300,000 new cases yearly. The five-year survival rate is approximately 50% and the major challenges to improving patient prognosis include late presentation, treatment resistance, second primary tumours and the lack of targeted therapies. Therefore, there is a compelling need to develop novel therapeutic strategies. In this study, we have examined the effect of lysophosphatidic acid (LPA) on OSCC cell migration, invasion and response to radiation, and investigated the contribution of cyclooxygenase-2 (COX-2) in mediating the tumour promoting effects of LPA. Using the TCGA data set, we show that the expression of the lipid phosphate phosphatases (LPP), LPP1 and LPP3, was significantly down-regulated in OSCC tissues. There was no significant difference in the expression of the ENPP2 gene, which encodes for the enzyme autotaxin (ATX) that produces LPA, between OSCCs and control tissues but ENPP2 levels were elevated in a subgroup of OSCCs. To explore the phenotypic effects of LPA, we treated OSCC cell lines with LPA and showed that the lipid enhanced migration and invasion as well as suppressed the response of the cells to irradiation. We also show that LPA increased COX-2 mRNA and protein levels in OSCC cell lines and inhibition of COX-2 activity with the COX-2 inhibitor, NS398, attenuated LPA-induced OSCC cell migration. Collectively, our data show for the first time that COX-2 mediates some of the pro-tumorigenic effects of LPA in OSCC and identifies the ATX-LPP-LPA-COX-2 pathway as a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Mariati Abdul Rahman
- Department of Oral and Craniofacial Sciences, University of Malaya, Kuala Lumpur, Malaysia.,Department of Craniofacial Diagnostics and Biosciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - May Leng Tan
- Department of Oral and Craniofacial Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Robert J Hollows
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Wen Lin Chai
- Department of Restorative Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Jason P Mansell
- Department of Applied Sciences, University of the West of England, Bristol, United Kingdom
| | - Lee Fah Yap
- Department of Oral and Craniofacial Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Ian C Paterson
- Department of Oral and Craniofacial Sciences, University of Malaya, Kuala Lumpur, Malaysia.,Oral Cancer Research and Coordinating Centre, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Masi I, Caprara V, Bagnato A, Rosanò L. Tumor Cellular and Microenvironmental Cues Controlling Invadopodia Formation. Front Cell Dev Biol 2020; 8:584181. [PMID: 33178698 PMCID: PMC7593604 DOI: 10.3389/fcell.2020.584181] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
During the metastatic progression, invading cells might achieve degradation and subsequent invasion into the extracellular matrix (ECM) and the underlying vasculature using invadopodia, F-actin-based and force-supporting protrusive membrane structures, operating focalized proteolysis. Their formation is a dynamic process requiring the combined and synergistic activity of ECM-modifying proteins with cellular receptors, and the interplay with factors from the tumor microenvironment (TME). Significant advances have been made in understanding how invadopodia are assembled and how they progress in degradative protrusions, as well as their disassembly, and the cooperation between cellular signals and ECM conditions governing invadopodia formation and activity, holding promise to translation into the identification of molecular targets for therapeutic interventions. These findings have revealed the existence of biochemical and mechanical interactions not only between the actin cores of invadopodia and specific intracellular structures, including the cell nucleus, the microtubular network, and vesicular trafficking players, but also with elements of the TME, such as stromal cells, ECM components, mechanical forces, and metabolic conditions. These interactions reflect the complexity and intricate regulation of invadopodia and suggest that many aspects of their formation and function remain to be determined. In this review, we will provide a brief description of invadopodia and tackle the most recent findings on their regulation by cellular signaling as well as by inputs from the TME. The identification and interplay between these inputs will offer a deeper mechanistic understanding of cell invasion during the metastatic process and will help the development of more effective therapeutic strategies.
Collapse
Affiliation(s)
- Ilenia Masi
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Valentina Caprara
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Bagnato
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Rosanò
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome, Italy.,Institute of Molecular Biology and Pathology, CNR, Rome, Italy
| |
Collapse
|
9
|
Chang Z, Zhang P, Zhang M, Jun F, Hu Z, Yang J, Wu Y, Zhou R. Aloperine suppresses human pulmonary vascular smooth muscle cell proliferation via inhibiting inflammatory response. CHINESE J PHYSIOL 2019; 62:157-165. [PMID: 31535631 DOI: 10.4103/cjp.cjp_27_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Abnormal pulmonary arterial vascular smooth muscle cells (PASMCs) proliferation is critical pathological feature of pulmonary vascular remodeling that acts as driving force in the initiation and development of pulmonary arterial hypertension (PAH), ultimately leading to pulmonary hypertension. Aloperine is a main active alkaloid extracted from the traditional Chinese herbal Sophora alopecuroides and possesses outstanding antioxidation and anti-inflammatory effects. Our group found Aloperine has protective effects on monocroline-induced pulmonary hypertension in rats by inhibiting oxidative stress in previous researches. However, the anti-inflammation effects of Aloperine on PAH remain unclear. Therefore, to further explore whether the beneficial role of Aloperine on PAH was connected with its anti-inflammatory effects, we performed experiments in vitro. Aloperine significantly inhibited the proliferation and DNA synthesis of human pulmonary artery smooth muscle cells (HPASMCs) induced by platelet-derived growth factor-BB, blocked progression through G0/G1to S phase of the cell cycle and promoted total ratio of apoptosis. In summary, these results suggested that Aloperine negatively regulated nuclear factor-κB signaling pathway activity to exert protective effects on PAH and suppressed HPASMCs proliferation therefore has a potential value in the treatment of pulmonary hypertension by negatively modulating pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Zhi Chang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Peng Zhang
- General Hospital of Ningxia Medical University, Yinchuan 750004, PR China
| | - Min Zhang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Feng Jun
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Zhiqiang Hu
- General Hospital of Ningxia Medical University, Yinchuan 750004, PR China
| | - Jiamei Yang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Yuhua Wu
- General Hospital of Ningxia Medical University, Yinchuan 750004, PR China
| | - Ru Zhou
- Department of Pharmacology, College of Pharmacy; Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education; Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan 750004, PR China
| |
Collapse
|
10
|
Zhang M, Chang Z, Zhang P, Jing Z, Yan L, Feng J, Hu Z, Xu Q, Zhou W, Ma P, Hao Y, Zhou R. Protective effects of 18β-glycyrrhetinic acid on pulmonary arterial hypertension via regulation of Rho A/Rho kinsase pathway. Chem Biol Interact 2019; 311:108749. [DOI: 10.1016/j.cbi.2019.108749] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/26/2019] [Accepted: 07/15/2019] [Indexed: 11/28/2022]
|
11
|
Modulation of LPA1 receptor-mediated neuronal apoptosis by Saikosaponin-d: A target involved in depression. Neuropharmacology 2019; 155:150-161. [DOI: 10.1016/j.neuropharm.2019.05.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/13/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022]
|
12
|
Zhang X, Hwang YS. Cancer-associated fibroblast stimulates cancer cell invasion in an interleukin-1 receptor (IL-1R)-dependent manner. Oncol Lett 2019; 18:4645-4650. [PMID: 31611973 PMCID: PMC6781746 DOI: 10.3892/ol.2019.10784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/05/2019] [Indexed: 12/30/2022] Open
Abstract
Tumor microenvironment serves an important role in tumor growth and metastasis. Cancer cells can promote growth and malignancy by altering the surrounding stroma. Cancer-associated fibroblast (CAF) are an abundant cell type present within the tumor microenvironment and provide tumorigenic features by secreting cytokines. In the current study, the CAF-mediated invasion of oral squamous cell carcinoma (OSCC) was investigated and the associated mechanisms were elucidated. Cancer invasion was estimated using a Matrigel-coated Transwell chamber and FITC-gelatin matrix. To verify the effect of the tumor microenvironment, conditioned media (CM) from normal fibroblast (NF) and CAFs were prepared. An ELISA was performed to estimate the level of IL-1β. A proteome profiler human protease array was performed to verify the proteases affected by stimulation with CM, from CAF. Recombinant IL-1β protein increased the invasion of OSCC cells. IL-1β expression was higher in CAF than NF. CM from CAF (CM-CAF) increased cancer invasion and FITC-gelatin matrix degradation. The invasive capacity provided by CAF was abrogated by an IL-1 receptor (IL-1R) antagonist. Additionally, CM-CAF increased the secretion of ADAM 9 and Kallikrein 11 from OSCC cells. The invasion activity by CM-CAF was partially abrogated by the neutralization of ADAM 9 or Kallikrein 11. In conclusion, by providing stromal factor, CAFs were a critical inducer of OSCC invasion, and CAF secretes the required amount of IL-1β to increase cancer invasion activity. The invasive capacity of CAF was identified to be IL-1R-dependent. ADAM 9 and Kallikrein 11 were influencing factors involved in the increase of CAF-mediated cancer invasion.
Collapse
Affiliation(s)
- Xianglan Zhang
- Department of Pathology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China.,Oral Cancer Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Young Sun Hwang
- Department of Dental Hygiene, College of Health Science, Eulji University, Seongnam, Gyunggi-Do 13135, Republic of Korea
| |
Collapse
|
13
|
Ubiquitination and Long Non-coding RNAs Regulate Actin Cytoskeleton Regulators in Cancer Progression. Int J Mol Sci 2019; 20:ijms20122997. [PMID: 31248165 PMCID: PMC6627692 DOI: 10.3390/ijms20122997] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022] Open
Abstract
Actin filaments are a major component of the cytoskeleton in eukaryotic cells and play an important role in cancer metastasis. Dynamics and reorganization of actin filaments are regulated by numerous regulators, including Rho GTPases, PAKs (p21-activated kinases), ROCKs (Rho-associated coiled-coil containing kinases), LIMKs (LIM domain kinases), and SSH1 (slingshot family protein phosphate 1). Ubiquitination, as a ubiquitous post-transcriptional modification, deceases protein levels of actin cytoskeleton regulatory factors and thereby modulates the actin cytoskeleton. There is increasing evidence showing cytoskeleton regulation by long noncoding RNAs (lncRNAs) in cancer metastasis. However, which E3 ligases are activated for the ubiquitination of actin-cytoskeleton regulators involved in tumor metastasis remains to be fully elucidated. Moreover, it is not clear how lncRNAs influence the expression of actin cytoskeleton regulators. Here, we summarize physiological and pathological mechanisms of lncRNAs and ubiquitination control mediators of actin cytoskeleton regulators which that are involved in tumorigenesis and tumor progression. Finally, we briefly discuss crosstalk between ubiquitination and lncRNA control mediators of actin-cytoskeleton regulators in cancer.
Collapse
|
14
|
Genc GE, Hipolito VEB, Botelho RJ, Gumuslu S. Lysophosphatidic acid represses autophagy in prostate carcinoma cells. Biochem Cell Biol 2018; 97:387-396. [PMID: 30403494 DOI: 10.1139/bcb-2018-0164] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a small signaling phospholipid that mediates diverse functions including cell proliferation, migration, and survival by engaging LPA-agonized G-protein coupled receptors. Autophagy is a survival mechanism in response to nutrient depletion or organellar damage that encloses idle or damaged organelles within autophagosomes that are then delivered to lysosomes for degradation. However, the relationship between LPA and autophagy is largely unknown. The purpose of this study is to elucidate whether LPA affects autophagy through the ERK1/2 and (or) the Akt-mTOR signaling pathways. In this study, we investigated the effect of LPA on autophagy-regulating pathways in various prostate-derived cancer cells including PC3, LNCaP, and Du145 cells grown in complete medium and exposed to serum-free medium. Using Western blotting and ELISA, we determined that LPA stimulates the ERK and mTOR pathways in complete and serum-free medium. The mTOR pathway led to phosphorylation of S6K and ULK, which respectively stimulates protein synthesis and arrests autophagy. Consistent with this, LPA exposure suppressed autophagy as measured by LC3 maturation and formation of GFP-LC3 puncta. Altogether, these results suggest that LPA suffices to activate mTORC1 and suppress autophagy in prostate cancer cells.
Collapse
Affiliation(s)
- Gizem E Genc
- a Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Victoria E B Hipolito
- b Department of Chemistry and Biology and the Graduate Program in Molecular Science, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Roberto J Botelho
- b Department of Chemistry and Biology and the Graduate Program in Molecular Science, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Saadet Gumuslu
- a Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| |
Collapse
|
15
|
Kahue CN, Jerrell RJ, Parekh A. Expression of human papillomavirus oncoproteins E6 and E7 inhibits invadopodia activity but promotes cell migration in HPV-positive head and neck squamous cell carcinoma cells. Cancer Rep (Hoboken) 2018; 1:e1125. [PMID: 32721084 PMCID: PMC7941430 DOI: 10.1002/cnr2.1125] [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] [Received: 03/08/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/30/2022] Open
Abstract
Background The rapid increase in the incidence of head and neck squamous cell carcinoma (HNSCC) is caused by high‐risk human papillomavirus (HPV) infections. The HPV oncogenes E6 and E7 promote carcinogenesis by disrupting signaling pathways that control survival and proliferation. Although these cancers are often diagnosed with metastases, the mechanisms that regulate their dissemination are unknown. Aims The aim of this study was to determine whether the HPV‐16 E6 and E7 oncogenes affected the invasive and migratory properties of HNSCC cells which promote their spread and metastasis. Methods and results Invasiveness was determined using invadopodia assays which allow for quantitation of extracellular matrix (ECM) degradation by invadopodia which are proteolytic membrane protrusions that facilitate invasion. Using cell lines and genetic manipulations, we found that HPV inhibited invadopodia activity in aggressive cell lines which was mediated by the E6 and E7 oncogenes. Given these findings, we also tested whether HPV caused differences in the migratory ability of HNSCC cells using Transwell assays. In contrast to our invadopodia results, we found no correlation between HPV status and cell migration; however, blocking the expression of the E6 and E7 oncoproteins in a HPV‐positive (HPV+) HNSCC cell line resulted in decreased migration. Conclusions Our data suggest that the E6 and E7 oncoproteins are negative regulators of invadopodia activity but may promote migration in HPV+ HNSCC cells. Despite the need for ECM proteolysis to penetrate most tissues, the unique structure of the head and neck tissues in which these cancers arise may facilitate the spread of migratory cancer cells without significant proteolytic ability.
Collapse
Affiliation(s)
- Charissa N Kahue
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel J Jerrell
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aron Parekh
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
16
|
Staal J, Beyaert R. Inflammation and NF-κB Signaling in Prostate Cancer: Mechanisms and Clinical Implications. Cells 2018; 7:E122. [PMID: 30158439 PMCID: PMC6162478 DOI: 10.3390/cells7090122] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer is a highly prevalent form of cancer that is usually slow-developing and benign. Due to its high prevalence, it is, however, still the second most common cause of death by cancer in men in the West. The higher prevalence of prostate cancer in the West might be due to elevated inflammation from metabolic syndrome or associated comorbidities. NF-κB activation and many other signals associated with inflammation are known to contribute to prostate cancer malignancy. Inflammatory signals have also been associated with the development of castration resistance and resistance against other androgen depletion strategies, which is a major therapeutic challenge. Here, we review the role of inflammation and its link with androgen signaling in prostate cancer. We further describe the role of NF-κB in prostate cancer cell survival and proliferation, major NF-κB signaling pathways in prostate cancer, and the crosstalk between NF-κB and androgen receptor signaling. Several NF-κB-induced risk factors in prostate cancer and their potential for therapeutic targeting in the clinic are described. A better understanding of the inflammatory mechanisms that control the development of prostate cancer and resistance to androgen-deprivation therapy will eventually lead to novel treatment options for patients.
Collapse
Affiliation(s)
- Jens Staal
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
17
|
Harper K, R. Lavoie R, Charbonneau M, Brochu-Gaudreau K, Dubois CM. The Hypoxic Tumor Microenvironment Promotes Invadopodia Formation and Metastasis through LPA1 Receptor and EGFR Cooperation. Mol Cancer Res 2018; 16:1601-1613. [DOI: 10.1158/1541-7786.mcr-17-0649] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/03/2018] [Accepted: 05/17/2018] [Indexed: 11/16/2022]
|
18
|
Meirson T, Gil-Henn H. Targeting invadopodia for blocking breast cancer metastasis. Drug Resist Updat 2018; 39:1-17. [PMID: 30075834 DOI: 10.1016/j.drup.2018.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/04/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
Dissemination of cancer cells from the primary tumor and their spread to distant sites of the body is the leading cause of mortality in metastatic cancer patients. Metastatic cancer cells invade surrounding tissues and blood vessels by forming F-actin-rich protrusions known as invadopodia, which degrade the extracellular matrix and enable invasion of tumor cells through it. Invadopodia have now been observed in vivo, and recent evidence demonstrates direct molecular links between assembly of invadopodia and cancer metastasis in both mouse models and in human patients. While significant progress has been achieved in the last decade in understanding the molecular mechanisms and signaling pathways regulating invadopodia formation and function, the application of this knowledge to development of prognostic and therapeutic approaches for cancer metastasis has not been discussed before. Here, we provide a detailed overview of current prognostic markers and tests for cancer metastasis and discuss their advantages, disadvantages, and their predicted efficiency. Using bioinformatic patient database analysis, we demonstrate, for the first time, a significant correlation between invadopodia-associated genes to breast cancer metastasis, suggesting that invadopodia could be used as both a prognostic marker and as a therapeutic target for blocking cancer metastasis. We include here a novel network interaction map of invadopodia-associated proteins with currently available inhibitors, demonstrating a central role for the recently identified EGFR-Pyk2-Src-Arg-cortactin invadopodial pathway, to which re-purposing of existent inhibitors could be used to block breast cancer metastasis. We then present an updated overview of current cancer-related clinical trials, demonstrating the negligible number of trials focusing on cancer metastasis. We also discuss the difficulties and complexity of performing cancer metastasis clinical trials, and the possible development of anti-metastasis drug resistance when using a prolonged preventive treatment with invadopodia inhibitors. This review presents a new perspective on invadopodia-mediated tumor invasiveness and may lead to the development of novel prognostic and therapeutic approaches for cancer metastasis.
Collapse
Affiliation(s)
- Tomer Meirson
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel.
| |
Collapse
|
19
|
Li C, Liu PP, Tang DD, Song R, Zhang YQ, Lei S, Wu SJ. Targeting the RhoA-ROCK pathway to regulate T-cell homeostasis in hypoxia-induced pulmonary arterial hypertension. Pulm Pharmacol Ther 2018; 50:111-122. [PMID: 29673911 DOI: 10.1016/j.pupt.2018.04.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/17/2018] [Accepted: 04/05/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Hypoxic pulmonary arterial hypertension (PAH) is a crippling disease with limited therapeutic methods. The imbalance of T helper 17 cell (Th17)/regulatory T cell (Treg) plays an important role in the development of Hypoxic PAH. However, whether targeting the ras homolog family member A-Rho kinase (RhoA-ROCK) pathway (activation and inhibition) by lysophosphatidic acid (LPA) and fasudil (FSD) regulate T-cell homeostasis in Hypoxic PAH remain unknown. OBJECTIVE To examine the effects of LPA and FSD on hypoxic pulmonary vascular remodeling and homeostasis of Th17/Treg cells in Hypoxic PAH. METHODS Rats were exposed to hypoxia (10 ± 0.5% O2) to induce Hypoxic PAH. The experiments consists of two parts. Forty rats were randomly divided into four groups (n = 10): normoxia group, normoxia + LPA group, hypoxia group and hypoxia + LPA group. Thirty rats were randomly divided into another three groups (n = 10): normoxia group, hypoxia group, and hypoxia + FSD group. Rats in normoxia + LPA group and hypoxia + LPA group were intraperitoneally injected 40 μg/kg LPA daily. Rats in hypoxia + FSD group were intraperitoneally injected 30 mg/kg fasudil daily. The effects of LPA and FSD on the development of hypoxic PAH and right ventricle (RV) hypertrophy, on pulmonary vascular remodeling, and on changes of Th17/Treg cells and levels of interleukin-17 (IL-17) and IL-10 were examined. RESULTS PAH and RV hypertrophy occurred in rats exposed to hypoxia. LPA exacerbated hypoxic pulmonary vascular remodeling and FSD inhibited it. LPA increased Th17/Treg imbalance in peripheral blood and spleen. However, after treatment with FSD, hypoxic PAH rats showed an obvious reduction of Th17 cells as well as an increase of Treg cells. LPA increased the expression of phosphorylated-signal transducer and activator of transcription 3 (p-STAT3) and reduced the p-STAT5 in peripheral blood and spleen in hypoxic PAH rats. The expression of p-STAT3 and p-STAT5 in hypoxic PAH rats treated with FSD showed opposite changes. LPA increased the expression of IL-17 and reduced the IL-10 in small intrapulmonary arteries and serum in hypoxic PAH. However, the expression of IL-17 and IL-10 in hypoxic PAH rats treated with FSD showed opposite changes. CONCLUSIONS Activation and inhibition of RhoA-ROCK pathway by LPA and FSD modulated the homeostasis of Th17/Treg cells via regulating STAT3/STAT5 phosphorylation in hypoxic PAH. Thus, Apart from influence of pulmonary vascular remodeling, regulation of Th17/Treg homeostasis by RhoA-ROCK pathway play a key role in hypoxic PAH.
Collapse
Affiliation(s)
- Cheng Li
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Research Unit of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China
| | - Ping-Ping Liu
- Department of Emergency, Hunan Children's Hospital, Changsha, Hunan 410007, PR China
| | - Dou-Dou Tang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Research Unit of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China
| | - Rong Song
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Research Unit of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China
| | - Yi-Qing Zhang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Research Unit of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China
| | - Si Lei
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Research Unit of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China
| | - Shang-Jie Wu
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Research Unit of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, No.139 Middle Renmin Road, Changsha, Hunan 410011, PR China.
| |
Collapse
|
20
|
Huang H, He Y, Zhang L, Xiang H, Li D, Liu W, Xu XT, Goodin S, Zhang K, Zheng X. Phenethyl isothiocyanate in combination with dibenzoylmethane inhibits the androgen-independent growth of prostate cancer cells. Food Funct 2018; 9:2398-2408. [DOI: 10.1039/c7fo01983a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study investigates the inhibitory effect of PEITC and DBM in combination on the progression of androgen-dependent VCaP prostate tumors to androgen independence.
Collapse
|
21
|
Wang BG, Li JS, Liu YF, Xu Q. MicroRNA-200b suppresses the invasion and migration of hepatocellular carcinoma by downregulating RhoA and circRNA_000839. Tumour Biol 2017; 39:1010428317719577. [PMID: 28695771 DOI: 10.1177/1010428317719577] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs could mediate the targeted coding gene and the targeted non-coding RNA to form endogenous competition, which have an important regulatory role in tumorigenesis of many types of cancer, including hepatocellular carcinoma. The goal of this study was to characterize the role of miR-200b in the pathogenesis of hepatocellular carcinoma. We identified miR-200b that was predicted to regulate RhoA and circ_000839. Our data establish that miR-200b is expressed at a relatively low level in hepatocellular carcinoma ( p < 0.001). RhoA and circ_000839 are expressed at a relatively high level in hepatocellular carcinoma ( p < 0.001, respectively). Our mechanistic data indicate that RhoA is a direct target of miR-200b ( p < 0.001), binding of which affects the expression of invasion and migration in hepatocellular carcinoma cell lines ( p < 0.05). And correlation analysis showed that miR-200b was inversely correlated with RhoA and circ_000839 ( p = 0.012, p = 0.002, respectively), while RhoA was positively correlated with circ_000839 ( p < 0.001). Taken together, our data suggest that miR-200b could mediate RhoA gene and circ_000839 to form endogenous competition. And this is a direction for the association study of miR-200b and RhoA in the future.
Collapse
Affiliation(s)
- Ben-Gang Wang
- 1 Hepatobiliary Surgery Department of General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jun-Shuai Li
- 1 Hepatobiliary Surgery Department of General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yong-Feng Liu
- 1 Hepatobiliary Surgery Department of General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qian Xu
- 2 Tumor Etiology and Screening Department of General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Davies MR, Lee L, Feeley BT, Kim HT, Liu X. Lysophosphatidic acid-induced RhoA signaling and prolonged macrophage infiltration worsens fibrosis and fatty infiltration following rotator cuff tears. J Orthop Res 2017; 35:1539-1547. [PMID: 27505847 PMCID: PMC5502767 DOI: 10.1002/jor.23384] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/05/2016] [Indexed: 02/04/2023]
Abstract
Previous studies have suggested that macrophage-mediated chronic inflammation is involved in the development of rotator cuff muscle atrophy and degeneration following massive tendon tears. Increased RhoA signaling has been reported in chronic muscle degeneration, such as muscular dystrophy. However, the role of RhoA signaling in macrophage infiltration and rotator muscle degeneration remains unknown. Using a previously established rat model of massive rotator cuff tears, we found RhoA signaling is upregulated in rotator cuff muscle following a massive tendon-nerve injury. This increase in RhoA expression is greatly potentiated by the administration of a potent RhoA activator, lysophosphatidic acid (LPA), and is accompanied by increased TNFα and TGF-β1 expression in rotator cuff muscle. Boosting RhoA signaling with LPA significantly worsened rotator cuff muscle atrophy, fibrosis, and fatty infiltration, accompanied with massive monocytic infiltration of rotator cuff muscles. Co-staining of RhoA and the tissue macrophage marker CD68 showed that CD68+ tissue macrophages are the dominant cell source of increased RhoA signaling in rotator cuff muscles after tendon tears. Taken together, our findings suggest that LPA-mediated RhoA signaling in injured muscle worsens the outcomes of atrophy, fibrosis, and fatty infiltration by increasing macrophage infiltraion in rotator cuff muscle. Clinically, inhibiting RhoA signaling may represent a future direction for developing new treatments to improve muscle quality following massive rotator cuff tears. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1539-1547, 2017.
Collapse
Affiliation(s)
- Michael R. Davies
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Lawrence Lee
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
| | - Brian T. Feeley
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Hubert T. Kim
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Xuhui Liu
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| |
Collapse
|
23
|
Bagnato A, Rosanò L. Endothelin-1 receptor drives invadopodia: Exploiting how β-arrestin-1 guides the way. Small GTPases 2016; 9:394-398. [PMID: 27690729 DOI: 10.1080/21541248.2016.1235526] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Metastatization is a complex multistep process requiring fine-tuned regulated cytoskeleton re-modeling, mediated by the cross-talk of actin with interacting partners, such as the Rho GTPases. Our expanding knowledge of invadopodia, small invasive membrane protrusions composed of a core of F-actin, actin regulators and actin-binding proteins, and hotspots for secretion of extracellular matrix (ECM) proteinases, contributes to clarify critical steps of the metastatic program. Growth factor receptors and their intermediate signaling molecules, along with matrix adhesion and rigidity, pH and hypoxia, act as drivers of cytoskeleton changes and invadopodia formation. We recently pro-posed a novel route map by which cancer cells regulates invadopodia dynamics supporting metastasis as response to the endothelin A receptor (ETAR), among the highly druggable G-protein coupled receptors in cancer. The metastatic behavior exhibited by ovarian cancer cells overe-xpressing ETAR is now explained by the interplay with β-arrestin1 (β-arr1), a scaffold protein acting as signal-integrating module of RhoC and cofilin signaling for specific invadopodia formation, accomplished by its interaction with a Rho guanine nucleotide exchange factor (GEF), PDZ-RhoGEF, in a G-protein independent manner. Here, we summarize this novel activation of the RhoC pathway from ETAR/β-arr1 signaling that may be exploited therapeutically and discuss new perspectives for future directions of investigations.
Collapse
Affiliation(s)
- Anna Bagnato
- a Preclinical Models and New Therapeutic Agents Unit, Translational Research Functional Departmental Area , Regina Elena National Cancer Institute , Rome , Italy
| | - Laura Rosanò
- a Preclinical Models and New Therapeutic Agents Unit, Translational Research Functional Departmental Area , Regina Elena National Cancer Institute , Rome , Italy
| |
Collapse
|
24
|
Suppression of NADPH Oxidase Activity May Slow the Expansion of Osteolytic Bone Metastases. Healthcare (Basel) 2016; 4:healthcare4030060. [PMID: 27571113 PMCID: PMC5041061 DOI: 10.3390/healthcare4030060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/11/2016] [Accepted: 08/22/2016] [Indexed: 12/28/2022] Open
Abstract
Lysophosphatidic acid (LPA), generated in the microenvironment of cancer cells, can drive the proliferation, invasion, and migration of cancer cells by activating G protein-coupled LPA receptors. Moreover, in cancer cells that have metastasized to bone, LPA signaling can promote osteolysis by inducing cancer cell production of cytokines, such as IL-6 and IL-8, which can stimulate osteoblasts to secrete RANKL, a key promoter of osteoclastogenesis. Indeed, in cancers prone to metastasize to bone, LPA appears to be a major driver of the expansion of osteolytic bone metastases. Activation of NADPH oxidase has been shown to play a mediating role in the signaling pathways by which LPA, as well as RANKL, promote osteolysis. In addition, there is reason to suspect that Nox4 activation is a mediator of the feed-forward mechanism whereby release of TGF-beta from bone matrix by osteolysis promotes expression of PTHrP in cancer cells, and thereby induces further osteolysis. Hence, measures which can down-regulate NADPH oxidase activity may have potential for slowing the expansion of osteolytic bone metastases in cancer patients. Phycocyanin and high-dose statins may have utility in this regard, and could be contemplated as complements to bisphosphonates or denosumab for the prevention and control of osteolytic lesions. Ingestion of omega-3-rich flaxseed or fish oil may also have potential for controlling osteolysis in cancer patients.
Collapse
|