1
|
Wang K, Nguyen T, Gao Y, Guo R, Fan C, Liao H, Li J, Chai J, Xu X, Gong Y, Chen X. Androcin 18-1, a novel scorpion-venom peptide, shows a potent antitumor activity against human U87 cells via inducing mitochondrial dysfunction. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 170:104137. [PMID: 38759703 DOI: 10.1016/j.ibmb.2024.104137] [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: 02/13/2024] [Revised: 05/04/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Scorpion venom is a potent natural source for antitumor drug development due to the multiple action modes of anticancer components. Although the sequence of Androcin 18-1 has been identified from the transcriptome profile of the scorpion venom Androctonus bicolor, its bioactivity remains unclear. In this study, we described the antitumor mechanism whereby Androcin 18-1 inhibits the proliferation and induces apoptosis by inducing cell membrane disruption, ROS accumulation, and mitochondrial dysfunction in human U87 glioblastoma cells. Moreover, Androcin 18-1 could suppress cell migration via the mechanisms associated with cytoskeleton disorganization and MMPs/TIMPs expression regulation. The discovery of this work highlights the potential application of Androcin 18-1 in drug development for glioblastoma treatment.
Collapse
Affiliation(s)
- Kai Wang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Tienthanh Nguyen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Yihan Gao
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Ruiyin Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Chaofan Fan
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Hang Liao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Jiali Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Jinwei Chai
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Xueqing Xu
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China.
| | - Yuxin Gong
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
| |
Collapse
|
2
|
Galkina SI, Golenkina EA, Fedorova NV, Ksenofontov AL, Serebryakova MV, Stadnichuk VI, Baratova LA, Sud'ina GF. Effect of Dexamethasone on Adhesion of Human Neutrophils and Concomitant Secretion. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:2094-2106. [PMID: 38462453 DOI: 10.1134/s000629792312012x] [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: 07/26/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 03/12/2024]
Abstract
Neutrophils play a dual role in protecting the body. They are able to penetrate infected tissues and destroy pathogens there by releasing aggressive bactericidal substances. While into the surrounding tissues, the aggressive products secreted by neutrophils initiate development of inflammatory processes. Invasion of neutrophils into tissues is observed during the development of pneumonia in the patients with lung diseases of various etiologies, including acute respiratory distress syndrome caused by coronavirus disease. Synthetic corticosteroid hormone dexamethasone has a therapeutic effect in treatment of lung diseases, including reducing mortality in the patients with severe COVID-19. The acute (short-term) effect of dexamethasone on neutrophil adhesion to fibrinogen and concomitant secretion was studied. Dexamethasone did not affect either attachment of neutrophils to the substrate or their morphology. Production of reactive oxygen species (ROS) and nitric oxide (NO) by neutrophils during adhesion also did not change in the presence of dexamethasone. Dexamethasone stimulated release of metalloproteinases in addition to the proteins secreted by neutrophils during adhesion under control conditions, and selectively stimulated release of free amino acid hydroxylysine, a product of lysyl hydroxylase. Metalloproteinases play a key role and closely interact with lysyl hydroxylase in the processes of modification of the extracellular matrix. Therapeutic effect of dexamethasone could be associated with its ability to reorganize extracellular matrix in the tissues by changing composition of the neutrophil secretions, which could result in the improved gas exchange in the patients with severe lung diseases.
Collapse
Affiliation(s)
- Svetlana I Galkina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - Ekaterina A Golenkina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Natalia V Fedorova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexander L Ksenofontov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Marina V Serebryakova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | | | - Ludmila A Baratova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Galina F Sud'ina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
3
|
Becker KN, Pettee KM, Sugrue A, Reinard KA, Schroeder JL, Eisenmann KM. The Cytoskeleton Effectors Rho-Kinase (ROCK) and Mammalian Diaphanous-Related (mDia) Formin Have Dynamic Roles in Tumor Microtube Formation in Invasive Glioblastoma Cells. Cells 2022; 11:1559. [PMID: 35563863 PMCID: PMC9103681 DOI: 10.3390/cells11091559] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/21/2022] [Accepted: 04/30/2022] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma (GBM) is a progressive and lethal brain cancer. Malignant control of actin and microtubule cytoskeletal mechanics facilitates two major GBM therapeutic resistance strategies-diffuse invasion and tumor microtube network formation. Actin and microtubule reorganization is controlled by Rho-GTPases, which exert their effects through downstream effector protein activation, including Rho-associated kinases (ROCK) 1 and 2 and mammalian diaphanous-related (mDia) formins (mDia1, 2, and 3). Precise spatial and temporal balancing of the activity between these effectors dictates cell shape, adhesion turnover, and motility. Using small molecules targeting mDia, we demonstrated that global agonism (IMM02) was superior to antagonism (SMIFH2) as anti-invasion strategies in GBM spheroids. Here, we use IDH-wild-type GBM patient-derived cell models and a novel semi-adherent in vitro system to investigate the relationship between ROCK and mDia in invasion and tumor microtube networks. IMM02-mediated mDia agonism disrupts invasion in GBM patient-derived spheroid models, in part by inducing mDia expression loss and tumor microtube network collapse. Pharmacological disruption of ROCK prevented invasive cell-body movement away from GBM spheres, yet induced ultralong, phenotypically abnormal tumor microtube formation. Simultaneously targeting mDia and ROCK did not enhance the anti-invasive/-tumor microtube effects of IMM02. Our data reveal that targeting mDia is a viable GBM anti-invasion/-tumor microtube networking strategy, while ROCK inhibition is contraindicated.
Collapse
Affiliation(s)
- Kathryn N. Becker
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (K.N.B.); (K.M.P.)
| | - Krista M. Pettee
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (K.N.B.); (K.M.P.)
| | - Amanda Sugrue
- Department of Chemistry and Biochemistry, University of Heidelberg, Tiffin, OH 44883, USA;
| | - Kevin A. Reinard
- Division of Neurosurgery, ProMedica Toledo Hospital, Toledo, OH 43606, USA; (K.A.R.); (J.L.S.)
| | - Jason L. Schroeder
- Division of Neurosurgery, ProMedica Toledo Hospital, Toledo, OH 43606, USA; (K.A.R.); (J.L.S.)
- Department of Surgery, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Kathryn M. Eisenmann
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (K.N.B.); (K.M.P.)
| |
Collapse
|
4
|
Cardiovascular protection associated with cilostazol, colchicine and target of rapamycin inhibitors. J Cardiovasc Pharmacol 2022; 80:31-43. [PMID: 35384911 DOI: 10.1097/fjc.0000000000001276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/06/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT An alteration in extracellular matrix production by vascular smooth muscle cells is a crucial event in the pathogenesis of vascular diseases such as aging-related, atherosclerosis and allograft vasculopathy. The human target of rapamycin (TOR) is involved in the synthesis of extracellular matrix by vascular smooth muscle cells. TOR inhibitors reduce arterial stiffness, blood pressure, and left ventricle hypertrophy and decrease cardiovascular risk in kidney graft recipients and patients with coronary artery disease and heart allograft vasculopathy. Other drugs that modulate extracellular matrix production such as cilostazol and colchicine have also demonstrated a beneficial cardiovascular effect. Clinical studies have consistently shown that cilostazol confers cardiovascular protection in peripheral vascular disease, coronary artery disease, and cerebrovascular disease. In patients with type 2 diabetes, cilostazol prevents the progression of subclinical coronary atherosclerosis. Colchicine reduces arterial stiffness in patients with Familial Mediterranean Fever and patients with coronary artery disease. Pathophysiological mechanisms underlying the cardioprotective effect of these drugs may be related to interactions between the cytoskeleton, TOR signaling and cyclic AMP synthesis that remain to be fully elucidated. Adult vascular smooth muscle cells exhibit a contractile phenotype and produce little extracellular matrix. Conditions that upregulate extracellular matrix synthesis induce a phenotypic switch toward a synthetic phenotype. TOR inhibition with rapamycin reduces extracellular matrix production by promoting the change to the contractile phenotype. Cilostazol increases the cytosolic level of cyclic AMP, which in turn leads to a reduction in extracellular matrix synthesis. Colchicine is a microtubule-destabilizing agent that may enhance the synthesis of cyclic AMP.
Collapse
|
5
|
Yin Y, Zhang F, Zheng Z, Xiao Z, Yang Q, Gong N, Zhou J, Zuo D, Ai J. MMP-7 affects peritoneal ultrafiltration associated with elevated aquaporin-1 expression via MAPK/ERK pathway in peritoneal mesothelial cells. J Cell Mol Med 2021; 25:6887-6898. [PMID: 34117704 PMCID: PMC8278116 DOI: 10.1111/jcmm.16697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Peritoneal membrane dysfunction and the resulting ultrafiltration failure are the major disadvantages of long-term peritoneal dialysis (PD). It becomes increasingly clear that mesothelial cells play a vital role in the pathophysiological changes of the peritoneal membrane. Matrix metalloproteinases (MMPs) function in the extracellular environment of cells and mediate extracellular matrix turnover during peritoneal membrane homeostasis. We showed here that dialysate MMP-7 levels markedly increased in the patients with PD, and the elevated MMP-7 level was negatively associated with peritoneal ultrafiltration volume. Interestingly, MMP-7 could regulate the cell osmotic pressure and volume of human peritoneal mesothelial cells. Moreover, we provided the evidence that MMP-7 activated mitogen-activated protein kinases (MAPKs)-extracellular signal-regulated kinase 1/2 (ERK) pathway and subsequently promoted the expression of aquaporin-1 (AQP-1) resulting in the change of cell osmotic pressure. Using a specific inhibitor of ERK pathway abrogated the MMP-7-mediating AQP-1 up-regulation and cellular homeostasis. In summary, all the findings indicate that MMP-7 could modulate the activity of peritoneal cavity during PD, and dialysate MMP-7 might be a non-invasive biomarker and an alternative therapeutic target for PD patients with ultrafiltration failure.
Collapse
Affiliation(s)
- Yue Yin
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Fen Zhang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhuojun Zheng
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhiwen Xiao
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaomu Yang
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Nirong Gong
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia Zhou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Daming Zuo
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Ai
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Malone ET, Ganther S, Mena N, Radaic A, Shariati K, Kindberg A, Tafolla C, Kamarajan P, Fenno JC, Zhan L, Kapila YL. Treponema denticola-Induced RASA4 Upregulation Mediates Cytoskeletal Dysfunction and MMP-2 Activity in Periodontal Fibroblasts. Front Cell Infect Microbiol 2021; 11:671968. [PMID: 34094999 PMCID: PMC8171266 DOI: 10.3389/fcimb.2021.671968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
The periodontal complex consists of the periodontal ligament (PDL), alveolar bone, and cementum, which work together to turn mechanical load into biological responses that are responsible for maintaining a homeostatic environment. However oral microbes, under conditions of dysbiosis, may challenge the actin dynamic properties of the PDL in the context of periodontal disease. To study this process, we examined host-microbial interactions in the context of the periodontium via molecular and functional cell assays and showed that human PDL cell interactions with Treponema denticola induce actin depolymerization through a novel actin reorganization signaling mechanism. This actin reorganization mechanism and loss of cell adhesion is a pathological response characterized by an initial upregulation of RASA4 mRNA expression resulting in an increase in matrix metalloproteinase-2 activity. This mechanism is specific to the T. denticola effector protein, dentilisin, thereby uncovering a novel effect for Treponema denticola-mediated RASA4 transcriptional activation and actin depolymerization in primary human PDL cells.
Collapse
Affiliation(s)
- Erin Trent Malone
- Kapila Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| | - Sean Ganther
- Kapila Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| | - Nevina Mena
- Kapila Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| | - Allan Radaic
- Kapila Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| | - Keemia Shariati
- Kapila Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| | - Abigail Kindberg
- Bush Laboratory, Department of Cell and Tissue Biology, Biomedical Sciences Graduate, University of California San Francisco, San Francisco, CA, United States
| | - Christian Tafolla
- Kapila Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| | - Pachiyappan Kamarajan
- Kapila Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| | - J. Christopher Fenno
- Fenno Laboratory, Department of Biological and Material Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Ling Zhan
- Zhan Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| | - Yvonne L. Kapila
- Kapila Laboratory, Department of Orofacial Sciences, School of Dentistry San Francisco, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
7
|
Neutrophil Adhesion and the Release of the Free Amino Acid Hydroxylysine. Cells 2021; 10:cells10030563. [PMID: 33807594 PMCID: PMC7999338 DOI: 10.3390/cells10030563] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
During infection or certain metabolic disorders, neutrophils can escape from blood vessels, invade and attach to other tissues. The invasion and adhesion of neutrophils is accompanied and maintained by their own secretion. We have previously found that adhesion of neutrophils to fibronectin dramatically and selectively stimulates the release of the free amino acid hydroxylysine. The role of hydroxylysine and lysyl hydroxylase in neutrophil adhesion has not been studied, nor have the processes that control them. Using amino acid analysis, mass spectrometry and electron microscopy, we found that the lysyl hydroxylase inhibitor minoxidil, the matrix metalloproteinase inhibitor doxycycline, the PI3K/Akt pathway inhibitors wortmannin and the Akt1/2 inhibitor and drugs that affect the actin cytoskeleton significantly and selectively block the release of hydroxylysine and partially or completely suppress spreading of neutrophils. The actin cytoskeleton effectors and the Akt 1/2 inhibitor also increase the phenylalanine release. We hypothesize that hydroxylysine release upon adhesion is the result of the activation of lysyl hydroxylase in interaction with matrix metalloproteinase, the PI3K/Akt pathway and intact actin cytoskeleton, which play important roles in the recruitment of neutrophils into tissue through extracellular matrix remodeling.
Collapse
|
8
|
Nuclear factor-κB plays an important role in Tamarixetin-mediated inhibition of matrix metalloproteinase-9 expression. Eur J Pharmacol 2020; 893:173808. [PMID: 33345858 DOI: 10.1016/j.ejphar.2020.173808] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/29/2020] [Accepted: 12/07/2020] [Indexed: 01/20/2023]
Abstract
Flavonoids possess a broad spectrum of pharmacological properties, including anti-cancer, anti-oxidant and immunomodulatory activities. The current study explored the potential of some less-studied flavonoids in inhibiting Matrix Metalloproteinase-9 (MMP-9), a prominent biomarker, upregulated in a variety of cancers and known to promote migration and invasion of cancer cells. Amongst these, Tamarixetin, a naturally occurring flavonoid derivative of Quercetin, demonstrated significant dose-dependent inhibition of MMP-9 expression. Furthermore, a substantial inhibition of migration, invasion and clonogenic potential of HT1080 cells was also observed in the presence of Tamarixetin, which further suggests its role as a potential anti-cancer agent. It is noteworthy that Tamarixetin inhibits nuclear translocation as well the activity of nuclear factor kappa B (NFκB), both of which are functions essential for the activation of MMP-9 in promoting tumorigenesis. Additionally, the endogenous regulators of MMP-9 that tightly control its activity were also modulated by Tamarixetin, as evident from the 1.9 fold increase in the expression of Tissue Inhibitor of Metalloproteinase-1 (TIMP-1), with a concomitant 2.2 fold decrease in Matrix Metalloproteinase-14 (MMP-14) expression. The results obtained were further corroborated in three dimensional (3D) tumor models, which showed significant inhibition of MMP-9 activity as well as reduced invasive potential in the presence of Tamarixetin. Taken together, our observations demonstrate for the first time, the anti-invasive potential of Tamarixetin in cancer cells, indicating its possible use as a template for novel therapeutic applications.
Collapse
|
9
|
Antonosante A, Brandolini L, d’Angelo M, Benedetti E, Castelli V, Maestro MD, Luzzi S, Giordano A, Cimini A, Allegretti M. Autocrine CXCL8-dependent invasiveness triggers modulation of actin cytoskeletal network and cell dynamics. Aging (Albany NY) 2020; 12:1928-1951. [PMID: 31986121 PMCID: PMC7053615 DOI: 10.18632/aging.102733] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/02/2020] [Indexed: 06/10/2023]
Abstract
Glioblastoma (GB) is the most representative form of primary malignant brain tumour. Several studies indicated a pleiotropic role of CXCL8 in cancer due to its ability to modulate the tumour microenvironment, growth and aggressiveness of tumour cell. Previous studies indicated that CXCL8 by its receptors (CXCR1 and CXCR2) induced activation of the PI3K/p-Akt pathway, a crucial event in the regulation of cytoskeleton rearrangement and cell mobilization. Human GB primary cell culture and U-87MG cell line were used to study the effects of CXCR1 and CXCR2 blockage, by a dual allosteric antagonist, on cell migration and cytoskeletal dynamics. The data obtained point towards a specific effect of autocrine CXCL8 signalling on GB cell invasiveness by the activation of pathways involved in cell migration and cytoskeletal dynamics, such as PI3K/p-Akt/p-FAK, p-cortactin, RhoA, Cdc42, Acetylated α-tubulin and MMP2. All the data obtained support the concept that autocrine CXCL8 signalling plays a key role in the activation of an aggressive phenotype in primary glioblastoma cells and U-87MG cell line. These results provide new insights about the potential of a pharmacological approach targeting CXCR1/CXCR2 pathways to decrease migration and invasion of GB cells in the brain parenchyma, one of the principal mechanisms of recurrence.
Collapse
Affiliation(s)
- Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | | | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | | | - Sabino Luzzi
- San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | | |
Collapse
|
10
|
Palamà IE, D'Amone S, Ratano P, Donatelli A, Liscio A, Antonacci G, Testini M, Di Angelantonio S, Ragozzino D, Cortese B. Mechanical Durotactic Environment Enhances Specific Glioblastoma Cell Responses. Cancers (Basel) 2019; 11:E643. [PMID: 31075964 PMCID: PMC6562761 DOI: 10.3390/cancers11050643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/17/2019] [Accepted: 05/07/2019] [Indexed: 01/24/2023] Open
Abstract
Background: A hallmark of glioblastoma is represented by their ability to widely disperse throughout the brain parenchyma. The importance of developing new anti-migratory targets is critical to reduce recurrence and improve therapeutic efficacy. Methods: Polydimethylsiloxane substrates, either mechanically uniform or presenting durotactic cues, were fabricated to assess GBM cell morphological and dynamical response with and without pharmacological inhibition of NNMII contractility, of its upstream regulator ROCK and actin polymerization. Results: Glioma cells mechanotactic efficiency varied depending on the rigidity compliance of substrates. Morphologically, glioma cells on highly rigid and soft bulk substrates displayed bigger and elongated aggregates whereas on durotactic substrates the same cells were homogeneously dispersed with a less elongated morphology. The durotactic cues also induced a motility change, cell phenotype dependent, and with cells being more invasive on stiffer substrates. Pharmacological inhibition of myosin or ROCK revealed a rigidity-insensitivity, unlike inhibition of microfilament contraction and polymerization of F-actin, suggesting that alternative signalling is used to respond to durotactic cues. Conclusions: The presence of a distinct mechanical cue is an important factor in cell migration. Together, our results provide support for a durotactic role of glioma cells that acts through actomyosin contractility to regulate the aggressive properties of GBM cells.
Collapse
Affiliation(s)
| | - Stefania D'Amone
- National Research Council-Nanotechnology Institute, 73100 Lecce, Italy.
| | - Patrizia Ratano
- National Research Council-Nanotechnology Institute, 00185 Rome, Italy.
| | - Amato Donatelli
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy.
| | - Andrea Liscio
- National Research Council-Institute for Microelectronics and Microsystems, via del Fosso del Cavaliere 100, 00133 Roma, Italy.
| | - Giuseppe Antonacci
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, 00185 Rome, Italy.
| | | | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy.
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, 00185 Rome, Italy.
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy.
| | - Barbara Cortese
- National Research Council-Nanotechnology Institute, 00185 Rome, Italy.
| |
Collapse
|
11
|
Imai-Sumida M, Chiyomaru T, Majid S, Saini S, Nip H, Dahiya R, Tanaka Y, Yamamura S. Silibinin suppresses bladder cancer through down-regulation of actin cytoskeleton and PI3K/Akt signaling pathways. Oncotarget 2017; 8:92032-92042. [PMID: 29190895 PMCID: PMC5696161 DOI: 10.18632/oncotarget.20734] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 08/07/2017] [Indexed: 01/17/2023] Open
Abstract
Silibinin is the major active constituent of silymarin, an extract of milk thistle seeds. Silibinin has been shown to have significant anti-cancer effects in a variety of malignancies. However, the molecular mechanisms of silibinin action in bladder cancer have not been studied extensively. In the present study, we found that silibinin (10 μM) significantly suppressed proliferation, migration, invasion and induced apoptosis of T24 and UM-UC-3 human bladder cancer cells. Silibinin down-regulated the actin cytoskeleton and phosphatidylinositide 3-kinase (PI3K)/Akt signaling pathways in these cancer cell lines. These pathways were found to crosstalk through RAS cascades. We found that silibinin suppressed levels of trimethylated histone H3 lysine 4 and acetylated H3 at the KRAS promoter. Furthermore, silibinin targets long non-coding RNA: HOTAIR and ZFAS1, which are known to play roles as oncogenic factors in various cancers. This study shows that silibinin exerts anti-cancer effects through down-regulation of actin cytoskeleton and PI3K/Akt pathways and thus suppresses bladder cancer growth and progression.
Collapse
Affiliation(s)
- Mitsuho Imai-Sumida
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA, USA
| | - Takeshi Chiyomaru
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA, USA
- Current address: Department of Urology, National Hospital Organization Kagoshima Medical Center, Kagoshima, Japan
| | - Shahana Majid
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA, USA
| | - Sharanjot Saini
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA, USA
| | - Hannah Nip
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA, USA
| | - Rajvir Dahiya
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA, USA
| | - Yuichiro Tanaka
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA, USA
| | - Soichiro Yamamura
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA, USA
| |
Collapse
|
12
|
Mold Alkaloid Cytochalasin D Modifies the Morphology and Secretion of fMLP-, LPS-, or PMA-Stimulated Neutrophils upon Adhesion to Fibronectin. Mediators Inflamm 2017; 2017:4308684. [PMID: 28740333 PMCID: PMC5504967 DOI: 10.1155/2017/4308684] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/11/2017] [Accepted: 04/27/2017] [Indexed: 12/15/2022] Open
Abstract
Neutrophils play an essential role in innate immunity due to their ability to migrate into infected tissues and kill microbes with bactericides located in their secretory granules. Neutrophil transmigration and degranulation are tightly regulated by actin cytoskeleton. Invading pathogens produce alkaloids that cause the depolymerization of actin, such as the mold alkaloid cytochalasin D. We studied the effect of cytochalasin D on the morphology and secretion of fMLP-, LPS-, or PMA-stimulated human neutrophils upon adhesion to fibronectin. Electron microscopy showed that the morphology of the neutrophils adherent to fibronectin in the presence of various stimuli differed. But in the presence of cytochalasin D, all stimulated neutrophils exhibited a uniform nonspread shape and developed thread-like membrane tubulovesicular extensions (cytonemes) measuring 200 nm in diameter. Simultaneous detection of neutrophil secretory products by mass spectrometry showed that all tested stimuli caused the secretion of MMP-9, a key enzyme in the neutrophil migration. Cytochalasin D impaired the MMP-9 secretion but initiated the release of cathepsin G and other granular bactericides, proinflammatory agents. The release of bactericides apparently occurs through the formation, shedding, and lysis of cytonemes. The production of alkaloids which modify neutrophil responses to stimulation via actin depolymerization may be part of the strategy of pathogen invasion.
Collapse
|
13
|
Paiva KBS, Granjeiro JM. Matrix Metalloproteinases in Bone Resorption, Remodeling, and Repair. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:203-303. [PMID: 28662823 DOI: 10.1016/bs.pmbts.2017.05.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinases (MMPs) are the major protease family responsible for the cleavage of the matrisome (global composition of the extracellular matrix (ECM) proteome) and proteins unrelated to the ECM, generating bioactive molecules. These proteins drive ECM remodeling, in association with tissue-specific and cell-anchored inhibitors (TIMPs and RECK, respectively). In the bone, the ECM mediates cell adhesion, mechanotransduction, nucleation of mineralization, and the immobilization of growth factors to protect them from damage or degradation. Since the first description of an MMP in bone tissue, many other MMPs have been identified, as well as their inhibitors. Numerous functions have been assigned to these proteins, including osteoblast/osteocyte differentiation, bone formation, solubilization of the osteoid during bone resorption, osteoclast recruitment and migration, and as a coupling factor in bone remodeling under physiological conditions. In turn, a number of pathologies, associated with imbalanced bone remodeling, arise mainly from MMP overexpression and abnormalities of the ECM, leading to bone osteolysis or bone formation. In this review, we will discuss the functions of MMPs and their inhibitors in bone cells, during bone remodeling, pathological bone resorption (osteoporosis and bone metastasis), bone repair/regeneration, and emergent roles in bone bioengineering.
Collapse
Affiliation(s)
- Katiucia B S Paiva
- Laboratory of Extracellular Matrix Biology and Cellular Interaction (LabMec), Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| | - José M Granjeiro
- National Institute of Metrology, Quality and Technology (InMetro), Bioengineering Laboratory, Duque de Caxias, RJ, Brazil; Fluminense Federal University, Dental School, Niterói, RJ, Brazil
| |
Collapse
|
14
|
Bildyug N. Matrix metalloproteinases: an emerging role in regulation of actin microfilament system. Biomol Concepts 2017; 7:321-329. [PMID: 27763882 DOI: 10.1515/bmc-2016-0022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are implicated in many physiological and pathological processes, including contraction, migration, differentiation, and proliferation. These processes all involve cell phenotype changes, known to be accompanied by reorganization of actin cytoskeleton. Growing evidence indicates a correlation between MMP activity and the dynamics of actin system, suggesting their mutual regulation. Here, data on the influence of MMPs on the actin microfilament system, on the one hand, and the dependence of MMP expression and activation on the organization of actin structures, on the other hand, are reviewed. The different mechanisms of putative actin-MMP regulation are discussed.
Collapse
|
15
|
Logun MT, Bisel NS, Tanasse EA, Zhao W, Gunasekera B, Mao L, Karumbaiah L. Glioma Cell Invasion is Significantly Enhanced in Composite Hydrogel Matrices Composed of Chondroitin 4- and 4,6-Sulfated Glycosaminoglycans. J Mater Chem B 2016; 4:6052-6064. [PMID: 28217304 DOI: 10.1039/c6tb01083k] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of astrocytoma accounting for a majority of primary malignant brain tumors in the United States. Chondroitin sulfate proteoglycans (CSPGs) and their glycosaminoglycan (GAG) side chains are key constituents of the brain extracellular matrix (ECM) implicated in promoting tumor invasion. However, the mechanisms by which sulfated CS-GAGs promote brain tumor invasion are currently unknown. We hypothesize that glioma cell invasion is triggered by the altered sulfation of CS-GAGs in the tumor extracellular environment, and that this is potentially mediated by independent mechanisms involving CXCL12/CXCR4 and LAR signaling respectively. This was tested in vitro by encapsulating the human glioma cell line U87MG-EGFP into monosulfated (4-sulfated; CS-A), composite (4 and 4,6-sulfated; CS-A/E), unsulfated hyaluronic acid (HA), and unsulfated agarose (AG; polysaccharide) hydrogels within microfluidics-based choice assays. Our results demonstrated the enhanced preferential cell invasion into composite hydrogels, when compared to other hydrogel matrices (p<0.05). Haptotaxis assays demonstrated the significantly (p<0.05) faster migration of U87MG-EGFP cells in CXCL12 containing CS-GAG hydrogels when compared to other hydrogel matrices containing the same chemokine concentration. This is likely due to the significantly (p<0.05) greater affinity of composite CS-GAGs to CXCL12 over other hydrogel matrices. Results from qRT-PCR assays further demonstrated the significant (p<0.05) upregulation of the chemokine receptor CXCR4, and the CSPG receptor LAR in glioma cells within CS-GAG hydrogels compared to control hydrogels. Western blot analysis of cell lysates derived from glioma cells encapsulated in different hydrogel matrices further corroborate qRT-PCR results, and indicate the presence of a potential variant of LAR that is selectively expressed only in glioma cells encapsulated in CS-GAG hydrogels. These results suggest that sulfated CS-GAGs may directly induce enhanced invasion and haptotaxis of glioma cells associated with aggressive brain tumors via distinct mechanisms.
Collapse
Affiliation(s)
- Meghan T Logun
- Regenerative Bioscience Center, ADS Complex, University of Georgia, Athens, Georgia
| | - Nicole S Bisel
- Regenerative Bioscience Center, ADS Complex, University of Georgia, Athens, Georgia
| | - Emily A Tanasse
- College of Engineering, Boise State University, Boise, Idaho
| | - Wujun Zhao
- Department of Chemistry, University of Georgia, Athens, Georgia
| | - Bhagya Gunasekera
- Regenerative Bioscience Center, ADS Complex, University of Georgia, Athens, Georgia
| | - Leidong Mao
- College of Engineering, University of Georgia, Athens, Georgia
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, ADS Complex, University of Georgia, Athens, Georgia
| |
Collapse
|
16
|
Ghaffari SH, Yousefi M, Dizaji MZ, Momeny M, Bashash D, Zekri A, Alimoghaddam K, Ghavamzadeh A. Arsenic Trioxide Induces Apoptosis and Incapacitates Proliferation and Invasive Properties of U87MG Glioblastoma Cells through a Possible NF-κB-Mediated Mechanism. Asian Pac J Cancer Prev 2016; 17:1553-64. [DOI: 10.7314/apjcp.2016.17.3.1553] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
17
|
Nighot P, Al-Sadi R, Rawat M, Guo S, Watterson DM, Ma T. Matrix metalloproteinase 9-induced increase in intestinal epithelial tight junction permeability contributes to the severity of experimental DSS colitis. Am J Physiol Gastrointest Liver Physiol 2015; 309:G988-G997. [PMID: 26514773 PMCID: PMC4683300 DOI: 10.1152/ajpgi.00256.2015] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/07/2015] [Indexed: 01/31/2023]
Abstract
Recent studies have implicated a pathogenic role for matrix metalloproteinases 9 (MMP-9) in inflammatory bowel disease. Although loss of epithelial barrier function has been shown to be a key pathogenic factor for the development of intestinal inflammation, the role of MMP-9 in intestinal barrier function remains unclear. The aim of this study was to investigate the role of MMP-9 in intestinal barrier function and intestinal inflammation. Wild-type (WT) and MMP-9(-/-) mice were subjected to experimental dextran sodium sulfate (DSS) colitis by administration of 3% DSS in drinking water for 7 days. The mouse colonic permeability was measured in vivo by recycling perfusion of the entire colon using fluorescently labeled dextran. The DSS-induced increase in the colonic permeability was accompanied by an increase in intestinal epithelial cell MMP-9 expression in WT mice. The DSS-induced increase in intestinal permeability and the severity of DSS colitis was found to be attenuated in MMP-9(-/-) mice. The colonic protein expression of myosin light chain kinase (MLCK) and phospho-MLC was found to be significantly increased after DSS administration in WT mice but not in MMP-9(-/-) mice. The DSS-induced increase in colonic permeability and colonic inflammation was attenuated in MLCK(-/-) mice and MLCK inhibitor ML-7-treated WT mice. The DSS-induced increase in colonic surface epithelial cell MLCK mRNA was abolished in MMP-9(-/-) mice. Lastly, increased MMP-9 protein expression was detected within the colonic surface epithelial cells in ulcerative colitis cases. These data suggest a role of MMP-9 in modulation of colonic epithelial permeability and inflammation via MLCK.
Collapse
Affiliation(s)
- Prashant Nighot
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Rana Al-Sadi
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Manmeet Rawat
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico;
| | - Shuhong Guo
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - D Martin Watterson
- Northwestern University Feinberg School of Medicine, Chicago, Illinois; and
| | - Thomas Ma
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico; Veterans Affairs Medical Center, Albuquerque, New Mexico
| |
Collapse
|
18
|
Qiu X, He X, Huang Q, Liu X, Sun G, Guo J, Yuan D, Yang L, Ban N, Fan S, Tao T, Wang D. Overexpression of CCT8 and its significance for tumor cell proliferation, migration and invasion in glioma. Pathol Res Pract 2015; 211:717-25. [PMID: 26304164 DOI: 10.1016/j.prp.2015.04.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 03/22/2015] [Accepted: 04/01/2015] [Indexed: 01/01/2023]
Abstract
Overexpression of chaperonin containing t-complex polypeptide 1 (TCP1), or CCT, has been reported in various classes of malignancies. However, little is known about the expression of t-complex protein subunits TCP1theta (CCT8) in gliomas. In this study, the expression of CCT8 protein was detected using blotting analysis and immunohistochemistry. CCT8 was found to be overexpressed in gliomas and to correlate with the WHO grade of gliomas. To further investigate the biological function of CCT8 in gliomas, CCT8-silenced U87 glioblastoma multiforme (GBM) and U251MG cells were constructed using a small interference RNA (siRNA) sequence. The knockdown effect of CCT8 on proliferation and invasion in these cells was analyzed using the CCK8, flow cytometry cycle, scratch, transwell invasion and fluorescence assays. Compared with the controls, the glioma cells expressing CCT8-siRNA exhibited a significantly decreased proliferation and invasion capacity, as well as a dysregulated cell cytoskeleton. This study showed that high CCT8 protein expression might be related to poor outcome of glioma, and that CCT8 regulates the proliferation and invasion of glioblastomas.
Collapse
Affiliation(s)
- Xiaojun Qiu
- Department of Neurosurgery, Affiliated Hospital, Nantong University, Nantong 226001, PR China
| | - Xiaojuan He
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Pathology, Medical College, Nantong University, Nantong 226001, PR China
| | - Qingfeng Huang
- Department of Neurosurgery, Affiliated Hospital, Nantong University, Nantong 226001, PR China
| | - Xianchen Liu
- Department of Neurosurgery, Affiliated Hospital, Nantong University, Nantong 226001, PR China
| | - Guan Sun
- Department of Neurosurgery, Affiliated Hospital, Nantong University, Nantong 226001, PR China
| | - Jun Guo
- Department of Neurosurgery, Affiliated Hospital, Nantong University, Nantong 226001, PR China
| | - Damin Yuan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Pathology, Medical College, Nantong University, Nantong 226001, PR China
| | - Lixiang Yang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Pathology, Medical College, Nantong University, Nantong 226001, PR China
| | - Na Ban
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Pathology, Medical College, Nantong University, Nantong 226001, PR China
| | - Shaochen Fan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Pathology, Medical College, Nantong University, Nantong 226001, PR China
| | - Tao Tao
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Pathology, Medical College, Nantong University, Nantong 226001, PR China.
| | - Donglin Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Pathology, Medical College, Nantong University, Nantong 226001, PR China.
| |
Collapse
|
19
|
Kirmse R, Otto H, Ludwig T. Interdependency of cell adhesion, force generation and extracellular proteolysis in matrix remodeling. J Cell Sci 2011; 124:1857-66. [PMID: 21558415 DOI: 10.1242/jcs.079343] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It is becoming increasingly evident that the micromechanics of cells and their environment determine cell fate and function as much as soluble molecular factors do. We hypothesized that extracellular matrix proteolysis by membrane type 1 matrix metalloproteinase (MT1-MMP) depends on adhesion, force generation and rigidity sensing of the cell. Melanoma cells (MV3 clone) stably transfected with MT1-MMP, or the empty vector as a control, served as the model system. α2β1 integrins (cell adhesion), actin and myosin II (force generation and rigidity sensing) were blocked by their corresponding inhibitors (α2β1 integrin antibodies, Cytochalasin D, blebbistatin). A novel, anisotropic matrix array of parallel, fluorescently labeled collagen-I fibrils was used. Cleavage and bundling of the collagen-I fibrils, and spreading and durotaxis of the cells on this matrix array could be readily discerned and quantified by a combined set-up for fluorescence and atomic force microscopy. In short, expression of the protease resulted in the generation of structural matrix defects, clearly indicated by gaps in the collagen lattice and loose fiber bundles. This key feature of matrix remodeling depended essentially on the functionality of α2β1 integrin, the actin filament network and myosin II motor activity. Interference with any of these negatively impacted matrix cleavage and three-dimensional matrix entanglement of cells.
Collapse
Affiliation(s)
- Robert Kirmse
- German Cancer Research Center Heidelberg (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
20
|
Mali RS, Zhang XM, Chintala SK. A decrease in phosphorylation of cAMP-response element-binding protein (CREBP) promotes retinal degeneration. Exp Eye Res 2011; 92:528-36. [PMID: 21459086 DOI: 10.1016/j.exer.2011.03.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 03/15/2011] [Accepted: 03/24/2011] [Indexed: 01/14/2023]
Abstract
Excitotoxicity, induced either by N-Methyl-d-aspartate (NMDA) or kainic acid (KA), promotes irreversible loss of retinal ganglion cells (RGCs). Although the intracellular signaling mechanisms underlying excitotoxic cell death are still unclear, recent studies on the retina indicate that NMDA promotes RGC death by increasing phosphorylation of cyclic AMP (cAMP) response element (CRE)-binding protein (CREBP), while studies on the central nervous system indicate that KA promotes neuronal cell death by decreasing phosphorylation of CREBP, suggesting that CREBP can elicit dual responses depending on the excitotoxic-agent. Interestingly, the role of CREBP in KA-mediated death of RGCs has not been investigated. Therefore, by using an animal model of excitotoxicity, the aim of this study was to investigate whether excitotoxicity induces RGC death by decreasing Ser(133)-CREBP in the retina. Death of RGCs was induced in CD-1 mice by an intravitreal injection of 20 nmoles of kainic acid (KA). Decrease in CREBP levels was determined by immunohistochemistry, western blot analysis, and electrophoretic mobility gel shift assays (EMSAs). Immunohistochemical analysis indicated that CREBP was constitutively expressed in the nuclei of cells both in the ganglion cell layer (GCL) and in the inner nuclear layer (INL) of CD-1 mice. At 6 h after KA injection, nuclear localization of Ser(133)-CREBP was decreased in the GCL. At 24 h after KA injection, Ser(133)-CREBP was decreased further in GCL and the INL, and a decrease in Ser(133)-CREBP correlated with apoptotic death of RGCs and amacrine cells. Western blot analysis indicated that KA decreased Ser(133)-CREBP levels in retinal protein extracts. EMSA assays indicated that KA also reduced the binding of Ser(133)-CREBP to CRE consensus oligonucleotides. In contrast, intravitreal injection of CNQX, a non-NMDA glutamate receptor antagonist, restored the KA-induced decrease in Ser(133)-CREBP both in the GCL and INL, and inhibited loss of RGCs and amacrine cells. These results, for the first time, suggest that KA promotes retinal degeneration by reducing phosphorylation of Ser(133)-CREBP in the retina.
Collapse
Affiliation(s)
- Raghuveer S Mali
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA
| | | | | |
Collapse
|
21
|
Gueye Y, Ferhat L, Sbai O, Bianco J, Ould-Yahoui A, Bernard A, Charrat E, Chauvin JP, Risso JJ, Féron F, Rivera S, Khrestchatisky M. Trafficking and secretion of matrix metalloproteinase-2 in olfactory ensheathing glial cells: A role in cell migration? Glia 2011; 59:750-70. [DOI: 10.1002/glia.21146] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 12/20/2010] [Indexed: 02/06/2023]
|
22
|
On the association of lipid rafts to the spectrin skeleton in human erythrocytes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1808:183-90. [PMID: 20807499 DOI: 10.1016/j.bbamem.2010.08.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 08/19/2010] [Accepted: 08/25/2010] [Indexed: 11/21/2022]
Abstract
Lipid rafts are local inhomogeneities in the composition of the plasma membrane of living cells, that are enriched in sphingolipids and cholesterol in a liquid-ordered state, and proteins involved in receptor-mediated signalling. Interactions between lipid rafts and the cytoskeleton have been observed in various cell types. They are isolated as a fraction of the plasma membrane that resists solubilization by nonionic detergents at 4°C (detergent-resistant membranes, DRMs). We have previously described that DRMs are anchored to the spectrin-based membrane skeleton in human erythrocytes and can be released by increasing the pH and ionic strength of the solubilization medium with sodium carbonate. It was unexplained why this carbonate treatment was necessary and why this requirement was not reported by other workers in this area. We show here that when contaminating leukocytes are present in erythrocyte preparations that are subjected to detergent treatment, the isolation of DRMs can occur without the requirement for carbonate treatment. This is due to the uncontrolled breakdown of erythrocyte membrane components by hydrolases that are released from contaminating neutrophils that lead to proteolytic disruption of the supramolecular assembly of the membrane skeleton. Results presented here corroborate the concept that DRMs are anchored to the membrane skeleton through electrostatic interactions that most likely involve the spectrin molecule.
Collapse
|
23
|
Johnson TR, Koul S, Kumar B, Khandrika L, Venezia S, Maroni PD, Meacham RB, Koul HK. Loss of PDEF, a prostate-derived Ets factor is associated with aggressive phenotype of prostate cancer: regulation of MMP 9 by PDEF. Mol Cancer 2010; 9:148. [PMID: 20550708 PMCID: PMC2904725 DOI: 10.1186/1476-4598-9-148] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 06/15/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prostate-derived Ets factor (PDEF) is expressed in tissues of high epithelial content including prostate, although its precise function has not been fully established. Conventional therapies produce a high rate of cure for patients with localized prostate cancer, but there is, at present, no effective treatment for intervention in metastatic prostate cancer. These facts underline the need to develop new approaches for early diagnosis of aggressive prostate cancer patients, and mechanism based anti-metastasis therapies that will improve the outlook for hormone-refractory prostate cancer. In this study we evaluated role of prostate-derived Ets factor (PDEF) in prostate cancer. RESULTS We observed decreased PDEF expression in prostate cancer cell lines correlated with increased aggressive phenotype, and complete loss of PDEF protein in metastatic prostate cancer cell lines. Loss of PDEF expression was confirmed in high Gleason Grade prostate cancer samples by immuno-histochemical methods. Reintroduction of PDEF profoundly affected cell behavior leading to less invasive phenotypes in three dimensional cultures. In addition, PDEF expressing cells had altered cell morphology, decreased FAK phosphorylation and decreased colony formation, cell migration, and cellular invasiveness. In contrast PDEF knockdown resulted in increased migration and invasion as well as clonogenic activity. Our results also demonstrated that PDEF downregulated MMP9 promoter activity, suppressed MMP9 mRNA expression, and resulted in loss of MMP9 activity in prostate cancer cells. These results suggested that loss of PDEF might be associated with increased MMP9 expression and activity in aggressive prostate cancer. To confirm results we investigated MMP9 expression in clinical samples of prostate cancer. Results of these studies show increased MMP9 expression correlated with advanced Gleason grade. Taken together our results demonstrate decreased PDEF expression and increased MMP9 expression during the transition to aggressive prostate cancer. CONCLUSIONS These studies demonstrate for the first time negative regulation of MMP9 expression by PDEF, and that PDEF expression was lost in aggressive prostate cancer and was inversely associated with MMP9 expression in clinical samples of prostate cancer. Based on these exciting results, we propose that loss of PDEF along with increased MMP9 expression should serve as novel markers for early detection of aggressive prostate cancer.
Collapse
Affiliation(s)
- Thomas R Johnson
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, Denver Veterans Administrative Medical Center, and University of Colorado Comprehensive Cancer Center, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
The G-protein-coupled formylpeptide receptor FPR confers a more invasive phenotype on human glioblastoma cells. Br J Cancer 2010; 102:1052-60. [PMID: 20197768 PMCID: PMC2844039 DOI: 10.1038/sj.bjc.6605591] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Background: The G-protein-coupled formylpeptide receptor (FPR) that mediates chemotaxis of phagocytic leucocytes induced by bacterial and host-derived chemotactic peptides is selectively expressed by highly malignant human gliomas and contributes to tumour growth and angiogenesis. As invasion of surrounding normal tissues is one of the important features of tumour malignancy, we investigated the function of FPR in the invasive behaviour of human glioblastoma cells. Methods: Cells (FPR+ and FPR−) were isolated by single-cell cloning from a human glioblastoma cell line U-87MG. The FPR expression was assayed by flow cytometry and reverse transcription PCR. The function of FPR was investigated by chemotaxis and calcium flux induced by FPR agonist fMLF. Tumour cell motility was assayed by a wound-healing model in vitro. The growth and invasive phenotype were observed with subcutaneous implantation of tumour cells in nude mice. Over-expression of FPR in FPR− cells was performed by transfection of a plasmid vector-containing human FPR gene. Results: One of the glioma clones F9 that expressed high level of FPR showed a more ‘motile’ phenotype in vitro as compared with a clone G3 without FPR expression. Although F9 and G3 clones both formed subcutaneous tumours in nude mice, only F9 tumours invaded surrounding mouse connective tissues. Over-expression of FPR in G3 clone (G3F) increased the cell motility in vitro and the capacity of the cells to form more rapidly growing and invasive tumours in nude mice. We further found that, in addition to supernatant of necrotic tumour cells, foetal calf serum and human serum used in culture media contained FPR agonist activity and increased the motility of FPR-expressing glioblastoma cells. Conclusion: The expression of FPR is responsible for increased motility of human glioblastoma cells and their formation of highly invasive tumours.
Collapse
|
25
|
Silibinin inhibits invasive properties of human glioblastoma U87MG cells through suppression of cathepsin B and nuclear factor kappa B-mediated induction of matrix metalloproteinase 9. Anticancer Drugs 2010; 21:252-60. [DOI: 10.1097/cad.0b013e3283340cd7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
26
|
Abstract
In gliomas, germline gene alterations play a significant role during malignant transformation of progenitor glial cells, at least for families with occurrence of multiple cancers or with specific hereditary cancer syndromes. Scientific evidence during the last few years has revealed several constitutive genetic abnormalities that may influence glioma formation. These germline abnormalities are manifested as either gene polymorphisms or hemizygous mutations of key regulatory genes that are involved either in DNA repair or in apoptosis. Such changes, among others, include hemizygous alterations of the neurofibromatosis 1 (NF1) and p53 genes that are involved in apoptotic pathways, and alterations in multiple DNA repair genes such as mismatch repair (MMR) genes, x-ray cross-complementary genes (XRCC), and O6-methylguanine-DNA methyltransferase (MGMT) genes. Subsequent cellular changes include somatic mutations in cell cycle regulatory genes and genes involved in angiogenesis and invasion, leading eventually to tumor formation in various stages. Future molecular diagnosis may identify new genomic regions that could harbor genes important for glioma predisposition and aid in the early diagnosis of these patients and genetic counseling of their families.
Collapse
Affiliation(s)
- Athanassios P Kyritsis
- University Hospital of Ioannina, Neurosurgical Research Institute, University of Ioannina School of Medicine, University Campus, Ioannina 45110, Greece.
| | | | | | | |
Collapse
|
27
|
Gondi CS, Rao JS. Therapeutic potential of siRNA-mediated targeting of urokinase plasminogen activator, its receptor, and matrix metalloproteinases. Methods Mol Biol 2009; 487:267-81. [PMID: 19301652 PMCID: PMC2677983 DOI: 10.1007/978-1-60327-547-7_13] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Targeting proteases and their activators would retard the invasive ability of cancer cells, and has been shown to induce apoptosis in certain instances. Various methods have been developed to specifically target protease molecules in an attempt to retard invasion and migration. Of these methods, RNA interference (RNAi) holds great therapeutic potential. RNAi technology is now being used to target specific molecules for use as potential anti-cancer agents. RNAi-mediated silencing is almost catalytic when compared to anti-sense silencing. Of these targets, the uPAR-uPA system and MMPs holds great promise. Targeting uPA/uPAR may provide additive or synergistic treatment benefits if used in combination with conventional therapeutics such as chemotherapy or radiation. Studies point to the fact that specifically targeting MMP-9 or MMP-2 singly or in combination with other proteases could have specific therapeutic implications in the treatment of cancer. In this chapter we discuss the therapeutic potential of siRNA-mediated targeting of the uPAR-uPA system and MMPs as therapeutic agents for the treatment of cancer.
Collapse
Affiliation(s)
- Christopher S. Gondi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL 61656, USA
| | - Jasti S. Rao
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL 61656, USA,Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL 61656, USA,Correspondence: JS Rao, Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Box-1649, Peoria, IL 61656, USA; (309) 671-3445, E-mail
| |
Collapse
|
28
|
Sbai O, Ferhat L, Bernard A, Gueye Y, Ould-Yahoui A, Thiolloy S, Charrat E, Charton G, Tremblay E, Risso JJ, Chauvin JP, Arsanto JP, Rivera S, Khrestchatisky M. Vesicular trafficking and secretion of matrix metalloproteinases-2, -9 and tissue inhibitor of metalloproteinases-1 in neuronal cells. Mol Cell Neurosci 2008; 39:549-68. [PMID: 18817873 DOI: 10.1016/j.mcn.2008.08.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 07/08/2008] [Accepted: 08/09/2008] [Indexed: 11/17/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are endopeptidases that cleave matrix, soluble and membrane-bound proteins and are regulated by their endogenous inhibitors the tissue inhibitors of MMPs (TIMPs). Nothing is known about MMP/TIMP trafficking and secretion in neuronal cells. We focussed our attention on the gelatinases MMP-2 and MMP-9, and their inhibitor TIMP-1. MMPs and TIMP-1 fused to GFP were expressed in N2a neuroblastoma and primary neuronal cells to study trafficking and secretion using real time video-microscopy, imaging, electron microscopy and biochemical approaches. We show that MMPs and TIMP-1 are secreted in 160-200 nm vesicles in a Golgi-dependent pathway. These vesicles distribute along microtubules and microfilaments, co-localise differentially with the molecular motors kinesin and myosin Va and undergo both anterograde and retrograde trafficking. MMP-9 retrograde transport involves the dynein/dynactin molecular motor. In hippocampal neurons, MMP-2 and MMP-9 vesicles are preferentially distributed in the somato-dendritic compartment and are found in dendritic spines. Non-transfected hippocampal neurons also demonstrate vesicular secretion of MMP-2 in both its pro- and active forms and gelatinolytic activity localised within dendritic spines. Our results show differential trafficking of MMP and TIMP-1-containing vesicles in neuronal cells and suggest that these vesicles could play a role in neuronal and synaptic plasticity.
Collapse
Affiliation(s)
- Oualid Sbai
- Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 6184 CNRS-Université de la Méditerranée, Faculté de Médecine, IFR Jean Roche, Bd Pierre Dramard, 13916 Marseille Cedex 20, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sinpitaksakul SN, Pimkhaokham A, Sanchavanakit N, Pavasant P. TGF-beta1 induced MMP-9 expression in HNSCC cell lines via Smad/MLCK pathway. Biochem Biophys Res Commun 2008; 371:713-8. [PMID: 18457660 DOI: 10.1016/j.bbrc.2008.04.128] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 04/25/2008] [Indexed: 10/22/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) plays roles in cancer progression by degrading the extracellular matrix and basement membrane. Many growth factors including Transforming growth factor-beta1 (TGF-beta1) could induce MMP-9 expression. We demonstrated that TGF-beta1 induced MMP-9 mRNA and protein in human head and neck squamous cell carcinoma cell lines. Application of TGF-beta receptor type I inhibitor (SB505124) reduced the MMP-9 expression markedly. Whilst, inhibitor of Myosin light chain kinase (MLCK) could reduce the level of secreted MMP-9 in both the supernatants and cell lysate but not the level of MMP-9 mRNA. These suggested that MLCK might regulate MMP-9 expression post-transcriptionally. Application of SB505124 and siRNA Smad2/3 reduced the phosphorylation of myosin light chain (MLC) suggested that MLC is downstream to TbetaRI/Smad2/3 signaling pathway. In conclusion, these results describe a novel mechanism for the potentiation of TGF-beta1 signaling to induce MMP-9 expression via Smad and MLCK.
Collapse
|
30
|
Samanna V, Ma T, Mak TW, Rogers M, Chellaiah MA. Actin polymerization modulates CD44 surface expression, MMP-9 activation, and osteoclast function. J Cell Physiol 2007; 213:710-20. [PMID: 17508356 DOI: 10.1002/jcp.21137] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CD44 and MMP-9 are implicated in cell migration. In the current study, we tested the hypothesis that actin polymerization is critical for CD44 surface expression and MMP-9 activity on the cell surface. To understand the underlying molecular mechanisms involved in CD44 surface expression and MMP-9 activity on the cell surface, osteoclasts were treated with bisphosphonate (BP) alendronate, cytochalasin D (Cyt D), and a broad-spectrum MMP inhibitor (GM6001). BP has been reported to block the mevalonate pathway, thereby preventing prenylation of small GTPase signaling required for actin cytoskeleton modulation. We show in this study that osteoclasts secrete CD44 and MMP-9 into the resorption bay during migration and bone resorption. Results indicate that actin polymerization is critical for CD44 surface expression and osteoclast function. In particular, the surface expression of CD44 and the membrane activity of MMP-9 are reduced in osteoclasts treated with alendronate and Cyt D despite the membrane levels of MMP-9 being unaffected. Although GM6001 blocked MMP-9 activity, osteoclast migration, and bone resorption, the surface levels of CD44 were unaffected. We suggest that the surface expression of CD44 requires actin polymerization. Disruption of podosome and actin ring structures by Cyt D and alendronate not only resulted in reduced localization of MMP-9 in these structures but also in osteoclast migration and bone resorption. These results suggest that inhibition of actin polymerization by alendronate and Cyt D is effective in blocking CD44/MMP-9 complex formation on the cell surface, secretion of active form of MMP-9, and osteoclast migration. CD44/MMP-9 complex formation may signify a unique motility-enhancing signal in osteoclast function.
Collapse
Affiliation(s)
- V Samanna
- Department of Biomedical Sciences, Dental School, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | | | | | |
Collapse
|
31
|
Gondi CS, Lakka SS, Dinh DH, Olivero WC, Gujrati M, Rao JS. Intraperitoneal injection of a hairpin RNA-expressing plasmid targeting urokinase-type plasminogen activator (uPA) receptor and uPA retards angiogenesis and inhibits intracranial tumor growth in nude mice. Clin Cancer Res 2007; 13:4051-60. [PMID: 17634529 PMCID: PMC2139987 DOI: 10.1158/1078-0432.ccr-06-3032] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE The purpose of this study was to evaluate the therapeutic potential of using plasmid-expressed RNA interference (RNAi) targeting urokinase-type plasminogen activator (uPA) receptor (uPAR) and uPA to treat human glioma. EXPERIMENTAL DESIGN In the present study, we have used plasmid-based RNAi to simultaneously down-regulate the expression of uPAR and uPA in SNB19 glioma cell lines and epidermal growth factor receptor (EGFR)--overexpressing 4910 human glioma xenografts in vitro and in vivo, and evaluate the i.p. route for RNAi-expressing plasmid administered to target intracranial glioma. RESULTS Plasmid-mediated RNAi targeting uPAR and uPA did not induce OAS1 expression as seen from reverse transcription-PCR analysis. In 4910 EGFR-overexpressing cells, down-regulation of uPAR and uPA induced the down-regulation of EGFR and vascular endothelial growth factor and inhibited angiogenesis in both in vitro and in vivo angiogenic assays. In addition, invasion and migration were inhibited as indicated by in vitro spheroid cell migration, Matrigel invasion, and spheroid invasion assays. We did not observe OAS1 expression in mice with preestablished intracranial tumors, which were given i.p. injections of plasmid-expressing small interfering RNA--targeting uPAR and uPA. Furthermore, the small interfering RNA plasmid targeting uPAR and uPA caused regression of preestablished intracranial tumors when compared with the control mice. CONCLUSION In conclusion, the plasmid-expressed RNAi targeting uPAR and uPA via the i.p. route has potential clinical applications for the treatment of glioma.
Collapse
Affiliation(s)
- Christopher S. Gondi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Sajani S. Lakka
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Dzung H. Dinh
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - William C. Olivero
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Meena Gujrati
- Department of Pathology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Jasti S Rao
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- *Correspondence to: J.S. Rao, Ph.D., Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, One Illini Drive, Peoria, IL 61605, USA; e-mail:
| |
Collapse
|
32
|
Mackenzie GG, Keen CL, Oteiza PI. Microtubules are required for NF-kappaB nuclear translocation in neuroblastoma IMR-32 cells: modulation by zinc. J Neurochem 2007; 99:402-15. [PMID: 17029595 DOI: 10.1111/j.1471-4159.2006.04005.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The relevance of a functional cytoskeleton for Nuclear Factor-kappaB (NF-kappaB) nuclear translocation was investigated in neuronal cells, using conditions that led to a disruption of the cytoskeleton [inhibition of tubulin (vinblastine, colchicine), or actin (cytochalasin D) polymerization and zinc deficiency]. We present evidence that an impairment in tubulin polymerization can inhibit the formation of the complex tubulin-dynein-karyopherin alpha-p50 that is required for neuronal retrograde and nuclear NF-kappaB transport. Cells treated with vinblastine, colchicine or cytochalasin D, and zinc deficient cells, all showed a low nuclear NF-kappaB binding activity, and low nuclear concentrations of RelA and p50. The altered nuclear translocation was reflected by a decreased transactivation of NF-kappaB-driven genes. The immunocytochemical characterization of cellular RelA showed that cytoskeleton disruption can lead to an altered distribution of RelA resulting in the formation of peripheral accumuli. These results support the concept that cytoskeleton integrity is necessary for the transport and translocation of NF-kappaB required for synapse to nuclei communication. We suggest that during development, as well as in the adult brain, conditions such as zinc deficiency, that affect the normal structure and function of the cytoskeleton can affect neuronal proliferation, differentiation, and survival by altering NF-kappaB nuclear translocation and subsequent impairment of NF-kappaB-dependent gene regulation.
Collapse
|
33
|
Abstract
Angiogenesis, the process by which new branches sprout from existing vessels, requires the degradation of the vascular basement membrane and remodeling of the ECM in order to allow endothelial cells to migrate and invade into the surrounding tissues. Serine, metallo, and cysteine proteinases are 3 types of a family of enzymes that proteolytically degrade various components of extracellular matrix. These proteases release various growth factors and also increase adhesive molecules and signaling pathway molecules upon their activation, which plays a significant role in angiogenesis. Downregulation of these molecules by antisense/siRNA or synthetic inhibitors decreases the levels of these molecules, inhibits the release of growth factors, and decreases the levels of various signaling pathway molecules, thereby leading to the inhibition of angiogenesis. Furthermore, MMPs degrade specific substrates and release angiogenic inhibitors which inhibit angiogenesis. Downregulation of 2 molecules, such as uPA and uPAR, uPAR and MMP-9, or Cathepsin B and MMP-9, are more effective to inhibit angiogenesis rather than downregulation of single molecules. However, careful testing of these combinations are most important because multiple effects of these combinations play a significant role in angiogenesis.
Collapse
Affiliation(s)
- Sajani S Lakka
- Division of Cancer Biology, Department of Biomedical and Therapeutic Sciences, University of Illinois College of Medicine-Peoria, Peoria, IL 61605, USA
| | | | | |
Collapse
|
34
|
Nico B, Corsi P, Ria R, Crivellato E, Vacca A, Roccaro AM, Mangieri D, Ribatti D, Roncali L. Increased matrix-metalloproteinase-2 and matrix-metalloproteinase-9 expression in the brain of dystrophic mdx mouse. Neuroscience 2006; 140:835-48. [PMID: 16650610 DOI: 10.1016/j.neuroscience.2006.02.077] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Revised: 01/31/2006] [Accepted: 02/27/2006] [Indexed: 11/29/2022]
Abstract
Brain edema and severe alterations of the glial and endothelial cells have recently been demonstrated in the dystrophin-deficient mdx mouse, an experimental model of Duchenne muscular dystrophy, and an increase in microvessel density in patients affected by Duchenne muscular dystrophy has also been shown. In order to further elucidate the mechanisms underlying the angiogenetic processes occurring in Duchenne muscular dystrophy, in this study we analyzed matrix-metalloproteinase-2 and -9 expression in the brain of 20-month-old mdx and control mice by means of immunohistochemistry, in situ hybridization, immunoblotting and gelatin zymography. Moreover, we studied vascular endothelial growth factor expression by means of Western blot and immunohistochemistry, and by dual immunofluorescence using anti-vascular endothelial growth factor and anti matrix-metalloproteinase-2 and-9 antibodies. Ultrastructural features of the brain choroidal plexuses were evaluated by electron microscopy. Spatial relationships between endothelium and astrocyte processes were studied by confocal laser microscopy, using an anti-CD31 antibody as a marker of endothelial cells, and anti-glial fibrillary acidic protein (GFAP) as a marker of glial cells. The results demonstrate that high expression of matrix-metalloproteinase-2 and matrix-metalloproteinase-9 protein content occurs in mdx brain and in choroidal plexuses where, by in situ hybridization, matrix-metalloproteinase-2 and matrix-metalloproteinase-9 mRNA was localized in the epithelial cells. Moreover, matrix-metalloproteinase-2 mRNA was found in both mdx perivascular astrocytes and blood vessels, while matrix-metalloproteinase-9 mRNA was localized in mdx vessels. Through zymography, increased expression of matrix-metalloproteinase-2 and matrix-metalloproteinase-9 was found in mdx brain compared with the controls. These enhanced matrix-metalloproteinase levels in mdx mice were found to be associated with increased vascular endothelial growth factor expression, as determined by immunoblotting and immunocytochemistry and with ultrastructural alterations of the mdx choroidal epithelial cells and brain vessels, as previously reported [Nico B, Frigeri A, Nicchia GP, Corsi P, Ribatti D, Quondamatteo F, Herken R, Girolamo F, Marzullo A, Svelto M, Roncali L (2003) Severe alterations of endothelial and glial cells in the blood-brain barrier of dystrophic mdx mice. Glia 42:235-251]. Indeed, in the mdx epithelial cells of the plexuses, the apical microvilli were located on the lateral membranes, whereas in the controls they were uniformly distributed over the free ventricular surface. Moreover, by dual immunofluorescence, a colocalization of vascular endothelial growth factor and matrix-metalloproteinase-2 and matrix-metalloproteinase-9 was found in the ependymal and epithelial cells of plexuses in mdx mice and, under confocal laser microscopy, mdx CD-31 positive vessels were enveloped by less GFAP-positive astrocyte processes than the controls. Overall, these data point to a specific pathogenetic role of matrix-metalloproteinase-2 and matrix-metalloproteinase-9 in neurological dysfunctions associated with Duchenne muscular dystrophy.
Collapse
MESH Headings
- Animals
- Astrocytes/enzymology
- Astrocytes/pathology
- Blood-Brain Barrier/enzymology
- Blood-Brain Barrier/pathology
- Blood-Brain Barrier/physiopathology
- Brain/enzymology
- Brain/pathology
- Brain/physiopathology
- Choroid Plexus/enzymology
- Choroid Plexus/pathology
- Disease Models, Animal
- Endothelial Cells/enzymology
- Endothelial Cells/pathology
- Ependyma/enzymology
- Ependyma/pathology
- Female
- Immunohistochemistry
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Microcirculation/enzymology
- Microcirculation/pathology
- Microcirculation/physiopathology
- Microscopy, Electron, Transmission
- Microvilli/enzymology
- Microvilli/pathology
- Muscular Dystrophy, Duchenne/enzymology
- Muscular Dystrophy, Duchenne/physiopathology
- Neovascularization, Pathologic/enzymology
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/physiopathology
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- RNA, Messenger/metabolism
- Up-Regulation/physiology
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- B Nico
- Department of Human Anatomy and Histology, University of Bari Medical School, Piazza Giulio Cesare, 11, Policlinico, I-70124 Bari, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Cell motility is an essential cellular process for a variety of biological events. The process of cell migration requires the integration and coordination of complex biochemical and biomechanical signals. The protrusion force at the leading edge of a cell is generated by the cytoskeleton, and this force generation is controlled by multiple signaling cascades. The formation of new adhesions at the front and the release of adhesions at the rear involve the outside-in and inside-out signaling mediated by integrins and other adhesion receptors. The traction force generated by the cell on the extracellular matrix (ECM) regulates cell-ECM adhesions, and the counter force exerted by ECM on the cell drives the migration. The polarity of cell migration can be amplified and maintained by the feedback loop between the cytoskeleton and cell-ECM adhesions. Cell migration in three-dimensional ECM has characteristics distinct from that on two-dimensional ECM. The migration of cells is initiated and modulated by external chemical and mechanical factors, such as chemoattractants and the mechanical forces acting on the cells and ECM, as well as the surface density, distribution, topography, and rigidity of the ECM.
Collapse
Affiliation(s)
- Song Li
- Department of Bioengineering and Center for Tissue Engineering, University of California, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
36
|
St-Pierre Y, Couillard J, Van Themsche C. Regulation of MMP-9 gene expression for the development of novel molecular targets against cancer and inflammatory diseases. Expert Opin Ther Targets 2005; 8:473-89. [PMID: 15469396 DOI: 10.1517/14728222.8.5.473] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The need to pharmacologically control the proteolytic activity of matrix metalloproteinases (MMPs) has been commonly acknowledged, despite its limited efficacy in clinical trials. Among the reasons that explain this failure is our limited understanding of the signals that control the expression of MMPs in different cell types during different pathological conditions. Thus, future therapies must rely on more selective approaches. With the continually increasing body of proof implicating MMPs in a large number of diseases, it has become a priority to establish the pertinence of molecules involved in the signalling pathways leading to the expression of these enzymes. MMP-9 is a case in point: its dramatic overexpression in cancer and various inflammatory conditions clearly points to the molecular mechanisms controlling its expression as a potential target for eventual rational therapeutic intervention. In this article, recent progress in the signalling pathways that regulate MMP-9 expression is reviewed, and the latest strategies to be considered in the search for a specific inhibitor of its expression are presented.
Collapse
Affiliation(s)
- Yves St-Pierre
- INRS-Institut Armand-Frappier, University of Quebec, 531 Boulevard des Prairies, Laval, Quebec, H7V 1B7, Canada.
| | | | | |
Collapse
|
37
|
Zhang W, Nwagwu C, Le DM, Yong VW, Song H, Couldwell WT. Increased invasive capacity of connexin43-overexpressing malignant glioma cells. J Neurosurg 2003; 99:1039-46. [PMID: 14705732 DOI: 10.3171/jns.2003.99.6.1039] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Malignant glioma cells, similar to astrocytes, express connexin43 (Cx43) universally but at widely varied levels. Data from previous studies have demonstrated that malignant glioma cells form functional gap junction channels among themselves as well as with astrocytes and that such a communication has the potential to modulate the phenotypic characteristics of astrocytes. Recently, gap junctions have been demonstrated to play a role in the invasive phenotype of malignant gliomas. In this study, the authors have further investigated the motility and invasion ability of Cx43-overexpressing and Cx43-deficient malignant glioma cells. METHODS Using a standard invasion system of a Matrigel transwell invasion chamber, the authors found that the number of Cx43-transfected C6 glioma cells (C6-Cx43 cells) migrating through the Matrigel-coated membrane was similar to that of mock-transfected control cells (C6-mock cells) during the first 24 hours, but increased significantly thereafter. When these cells were cocultured with astrocytes, the number of invading C6-Cx43 cells was more than threefold greater than the number of invading C6-mock cells. Results of an in vitro cell motility assay also demonstrated that C6-Cx43 cells were more motile and scatter-active than C6-mock cells. Furthermore, zymographic analysis of MMPs, an important determinant in glioma invasion, demonstrated that the amounts of MMP-2 and MMP-9 in culture medium collected from C6-Cx43 cells were orders of magnitude higher than those from C6-mock cells. In addition, BB-94, a synthetic MMP inhibitor, significantly inhibited C6-Cx43 cell invasion. CONCLUSIONS The overexpression of gap junction proteins in glioma cells and the intercellular communication between tumor and nontumor glia cells may play important roles in the facilitation of glioma cell invasion.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurosurgery, New York Medical College, Valhalla, New York, USA
| | | | | | | | | | | |
Collapse
|
38
|
Gondi CS, Lakka SS, Yanamandra N, Siddique K, Dinh DH, Olivero WC, Gujrati M, Rao JS. Expression of antisense uPAR and antisense uPA from a bicistronic adenoviral construct inhibits glioma cell invasion, tumor growth, and angiogenesis. Oncogene 2003; 22:5967-75. [PMID: 12955075 DOI: 10.1038/sj.onc.1206535] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2002] [Revised: 02/19/2003] [Accepted: 02/26/2003] [Indexed: 11/08/2022]
Abstract
Urokinase-type plasminogen activator (uPA) and its receptor (uPAR) play an important role in the invasiveness of gliomas and other infiltrative tumors. In glioma cell lines and tumors, high grade correlates with increased expression of uPAR and uPA. We report here the downregulation of uPAR and uPA by delivery of antisense sequences of uPAR and uPA in a single adenoviral vector, Ad-uPAR-uPA (Ad, adenovirus). The bicistronic construct (Ad-uPAR-uPA) infected glioblastoma cell line had significantly reduced levels of uPAR, uPA enzymatic activity and immunoreactivity for these proteins when compared to controls. The Ad-uPAR-uPA infected cells showed a markedly lower level of invasion in the Matrigel invasion assays, and their spheroids failed to invade the fetal rat brain aggregates in the coculture system. Intracranial injection of SNB19 cells with the Ad-uPAR-uPA antisense bicistronic construct showed inhibited invasiveness and tumorigenicity. Subcutaneous injections of bicistronic antisense constructs into established tumors (U87 MG) caused regression of those tumors. Our results support the therapeutic potential of targeting the individual components of the uPAR-uPA system by using a single adenovirus construct for the treatment of glioma and other invasive cancers.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Carcinogenicity Tests
- Cell Movement
- Central Nervous System Neoplasms/blood supply
- Central Nervous System Neoplasms/genetics
- Central Nervous System Neoplasms/pathology
- Central Nervous System Neoplasms/therapy
- DNA, Antisense/administration & dosage
- DNA, Antisense/genetics
- Gene Expression
- Genetic Therapy/methods
- Genetic Vectors/genetics
- Glioma/blood supply
- Glioma/genetics
- Glioma/pathology
- Glioma/therapy
- Humans
- Mice
- Mice, Nude
- Neoplasm Invasiveness
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/therapy
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Receptors, Urokinase Plasminogen Activator
- Tumor Cells, Cultured
- Urokinase-Type Plasminogen Activator/genetics
- Urokinase-Type Plasminogen Activator/immunology
- Urokinase-Type Plasminogen Activator/metabolism
Collapse
Affiliation(s)
- Christopher S Gondi
- Division of Cancer Biology, Department of Biomedical and Therapeutic Sciences, University of Illinois, Peoria, IL, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Chen NX, Geist DJ, Genetos DC, Pavalko FM, Duncan RL. Fluid shear-induced NFkappaB translocation in osteoblasts is mediated by intracellular calcium release. Bone 2003; 33:399-410. [PMID: 13678782 DOI: 10.1016/s8756-3282(03)00159-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Bone formation in response to exogenous mechanical loading is dependent on prostaglandin synthesis by the inducible isoform of cyclooxygenase, COX-2. While several transcription factors target the COX-2 gene, we examined the role of nuclear factor kappa B (NFkappaB) on COX-2 upregulation in osteoblasts in response to fluid shear due to its involvement in immune and inflammatory responses in other cell types. Application of 12 dyn/cm2 laminar flow to MC3T3-E1 osteoblast-like cells resulted in translocation of NFkappaB to the nucleus within 1 h of the onset of shear, with NFkappaB returning to the cytoplasm after 2 h of continuous flow. NFkappaB translocation in response to shear was inhibited by the protease inhibitor, Nalpha-p-tosyl-L-lysine chloromethylketone hydrochloride (TLCK), or a cell-permeant peptide that blocks the nuclear localization sequence (NLS) on NFkappaB. Block of NFkappaB translocation with these inhibitors blocked the shear-induced upregulation of COX-2. We found that disruption of the actin cytoskeleton with cytochalasin D or microtubules with nocodozol did not alter NFkappaB translocation in response to shear. However, addition of the intracellular Ca2+ chelator BAPTA completely blocked NFkappaB translocation. While block of Ca2+ entry with channel blockers failed to inhibit NFkappaB translocation, inhibition of phospholipase C (PLC)-induced intracellular Ca2+ release with the PLC inhibitor U73122 completely abrogated the NFkappaB response to shear. These data indicate that NFkappaB translocation to the nucleus is essential for the fluid shear-induced increase in COX-2. Further, these studies suggest that intracellular Ca2+ release, but not the cytoskeletal architecture, is important to NFkappaB translocation.
Collapse
Affiliation(s)
- Neal X Chen
- Department of Anatomy, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
40
|
Bauvois B, Puiffe ML, Bongui JB, Paillat S, Monneret C, Dauzonne D. Synthesis and biological evaluation of novel flavone-8-acetic acid derivatives as reversible inhibitors of aminopeptidase N/CD13. J Med Chem 2003; 46:3900-13. [PMID: 12930151 DOI: 10.1021/jm021109f] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The cell surface aminopeptidase N (APN/CD13), overexpressed in tumor cells, plays a critical role in angiogenesis. However, potent, selective, and, particularly, noncytotoxic inhibitors ot this protein are lacking, and the present work was undertaken with the aim of developing a new generation of noncytotoxic inhibitors that bind to APN/CD13. In this context, we have synthesized a series of novel flavone-8-acetic acid derivatives. Among the herein described and evaluated compounds, the 2',3-dinitroflavone-8-acetic acid (19b) proved to be the most efficient and exhibited an IC(50) of 25 microM which is 2.5 times higher than that of bestatin (1), the natural known inhibitor of APN/CD13. However, in contrast to bestatin (1), the dinitroflavone 19b did not induce any cytotoxicity to cultured human model cells. The presence of other substituents such as NO(2) or OCH(3) groups at the 3'- or 4'-position of the B phenyl group, or the existence of steric constraints (compounds 24 and 29), did not improve selectivity and potency. The flavone 19b affinity for APN/CD13 is not recovered with other proteases such as matrix metalloproteinase-9 (MMP-9), angiotensin converting enzyme (ACE/CD143), neutral endopeptidase (NEP/CD10), gamma-glutamyl transpeptidase (CD224), or the serine proteases dipeptidyl peptidase IV (DPPIV/CD26) or cathepsin G.
Collapse
Affiliation(s)
- Brigitte Bauvois
- Unité 365 INSERM, Institut Curie, Section de Recherche, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
The invasive nature of brain-tumour cells makes an important contribution to the ineffectiveness of current treatment modalities, as the remaining tumour cells inevitably infiltrate the surrounding normal brain tissue, which leads to tumour recurrence. Such local invasion remains an important cause of mortality and underscores the need to understand in more detail the mechanisms of tumour invasiveness. Several proteases influence the malignant characteristics of gliomas--could their inhibition prove to be a useful therapeutic strategy?
Collapse
Affiliation(s)
- Jasti S Rao
- Program of Cancer Biology, Department of Neurosurgery, University of Illinois College of Medicine-Peoria, 1 Illini Drive, Peoria, Illinois 61656, USA.
| |
Collapse
|
42
|
Gu Y, Xu YC, Wu RF, Nwariaku FE, Souza RF, Flores SC, Terada LS. p47phox participates in activation of RelA in endothelial cells. J Biol Chem 2003; 278:17210-7. [PMID: 12618429 DOI: 10.1074/jbc.m210314200] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Activation of endothelial cell NF-kappaB by interleukin (IL)-1 constitutes an event critical to the progression of the innate immune response. In this context, oxidants have been associated with NF-kappaB activation, although the molecular source and mechanism of targeting have remained obscure. We found that RelA, essential for NF-kappaB activation by IL-1, was associated with the NADPH oxidase adapter protein p47(phox) in yeast two-hybrid, coprecipitation, and in vitro binding studies. RelA and p47-GFP also colocalized in endothelial cells in focal submembranous dorsoventral protrusions. Overexpression of p47(phox) synergized with IL-1beta in the activation of an artificial kappaB-luciferase reporter and specifically augmented IL-1beta-induced RelA transactivation activity. p47(phox) overexpression also greatly increased IL-1beta-stimulated RelA phosphorylation, whereas it had no effect on I-kappaB degradation or on RelA nuclear translocation or kappaB binding. The tandem SH3 domains of p47(phox) were found to associate with a proline-rich mid-region of RelA (RelA-PR) located between the Rel homology and transactivation domains. The RelA-PR peptide blocked interaction of p47(phox) and RelA, and ectopic expression of RelA-PR abrogated IL-1beta-induced transactivation of the NF-kappaB-dependent E-selectin promoter. Further, suppression of NADPH oxidase function through the inhibitor diphenylene iodonium, the superoxide dismutase mimetic Mn(III) tetrakis(4-benzoic acid)porphyrin (MnTBAP), or expression of a dominant interfering mutant of a separate NADPH oxidase subunit (p67(V204A)) decreased IL-1beta-induced E-selectin promoter activation, suggesting that p47(phox) facilitates NF-kappaB activation through linkage with the NADPH oxidase. IL-1beta rapidly increased tyrosine phosphorylation of IL-1 type I receptor-associated proteins, suggesting that oxidants may operate through inactivation of local protein-tyrosine phosphatases in the proximal IL-1beta signaling pathway leading to RelA activation.
Collapse
Affiliation(s)
- Ying Gu
- Department of Internal Medicine, University of Texas Southwestern, Dallas, Texas 75216, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Tachado SD, Mayhew MW, Wescott GG, Foreman TL, Goodwin CD, McJilton MA, Terrian DM. Regulation of tumor invasion and metastasis in protein kinase C epsilon-transformed NIH3T3 fibroblasts. J Cell Biochem 2002; 85:785-97. [PMID: 11968018 DOI: 10.1002/jcb.10164] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Protein kinase C epsilon is an oncogenic, actin nucleating protein that coordinately regulates changes in cell growth and shape. Cells constitutively expressing PKCepsilon spontaneously acquire a polarized morphology and extend long cellular membrane protrusions. Here we report that the regulatory C1 domain of PKCepsilon contains an actin binding site that is essential for the formation of elongate invadopodial-like structures, increased pericellular metalloproteinase activity, in vitro invasion of a Matrigel barrier, and the invasion and metastasis of tumors grown in vivo by PKCepsilon-transformed NIH3T3 fibroblasts in nude mice. While removing this small actin binding motif caused a dramatic reversion of tumor invasion, the deletion mutant of PKCepsilon remained oncogenic and tumorigenic in this experimental system. We propose that PKCepsilon directly interacts with actin to stimulate polymerization and the extension of membrane protrusions that transformed NIH3T3 cells use in vivo to penetrate and degrade surrounding tissue boundaries.
Collapse
Affiliation(s)
- Souvenir D Tachado
- Department of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina 27858, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Sancéau J, Boyd DD, Seiki M, Bauvois B. Interferons inhibit tumor necrosis factor-alpha-mediated matrix metalloproteinase-9 activation via interferon regulatory factor-1 binding competition with NF-kappa B. J Biol Chem 2002; 277:35766-75. [PMID: 12105194 DOI: 10.1074/jbc.m202959200] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Enhanced expression of matrix metalloproteinase-9 (MMP-9) correlates with invasion during tumor progression. Interferons (IFNs) inhibit MMP-9 activation in response to tumor necrosis factor-alpha (TNF-alpha), and the latter activates the MMP-9 gene through NF-kappaB. Understanding the molecular basis for MMP-9 inhibition may provide tools to control cell invasion. The data reported here show the critical role of interferon regulatory factor-1 (IRF1) in the inhibition of MMP-9. (i) IFN treatment suppresses TNF-alpha-induced MMP-9 reporter activity in STAT1(+/+) cells but not in STAT1(-/-) cells. (ii) IRF1 transfection blocks TNF-alpha-mediated MMP-9 activation. (iii) IFNs phosphorylate STAT1 and induce IRF1 but do not affect Ikappa-B degradation nor NF-kappaB nuclear translocation. (iv) Nuclear NF-kappaB (p50/p65) and IRF1, but not STAT1, bind to the MMP-9 promoter region containing an IFN-responsive-like element overlapping the NF-kappaB-binding site. (v) Recombinant IRF1, although unable to bind to an NF-kappaB consensus sequence, competes with NF-kappaB proteins for binding to the MMP-9 promoter. (vi) Conversely recombinant p50/p65 proteins reduce IRF1-DNA binding. (vii) In cells cotransfected with IRF1 and/or p65 expression vectors, an excess of IRF1 reduces MMP-9 reporter activity, whereas an excess of p65 blocks the inhibitory effect of IFN-gamma. Thus, in contrast to the known synergism between IRF1 and NF-kappaB, our data identify a novel role for IRF1 as a competitive inhibitor of NF-kappaB binding to the particular MMP-9 promoter context.
Collapse
Affiliation(s)
- Josiane Sancéau
- Unité 365 INSERM, Section de Recherche, Institut Curie, 75248 Paris Cedex 05, France
| | | | | | | |
Collapse
|
45
|
Estève PO, Chicoine E, Robledo O, Aoudjit F, Descoteaux A, Potworowski EF, St-Pierre Y. Protein kinase C-zeta regulates transcription of the matrix metalloproteinase-9 gene induced by IL-1 and TNF-alpha in glioma cells via NF-kappa B. J Biol Chem 2002; 277:35150-5. [PMID: 12130632 DOI: 10.1074/jbc.m108600200] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulation of matrix metalloproteinase-9 (MMP-9) expression in glioma cells is one of the key processes in tumor invasion through the brain extracellular matrix. Although some studies have demonstrated the implication of classic protein kinase C (PKC) isoforms in the regulation of MMP-9 production by phorbol esters or lipopolysaccharide, the involvement of specific PKC isoforms in the signaling pathways leading to MMP-9 expression by inflammatory cytokines remains unclear. Here we report that the atypical PKC-zeta isoform participates in the induction of MMP-9 expression by interleukin-1 (IL-1) and tumor necrosis factor-alpha (TNF-alpha) in rat C6 glioma cells. Indeed, zymography and semi-quantitative reverse transcriptase-PCR analysis showed that pretreatment of C6 cells with PKC-zeta pseudosubstrate abolished MMP-9 activity and gene expression induced by IL-1 or TNF-alpha. Accordingly, IL-1 and TNF-alpha were able to induce PKC-zeta activity, as demonstrated by in vitro kinase assay using immunoprecipitated PKC-zeta. Furthermore, stable C6 clones overexpressing PKC-zeta, but not PKC-epsilon, displayed an up-regulation of MMP-9 constitutive expression as well as an increase of mmp-9 promoter activity. These processes were inhibited by an NF-kappaB-blocking peptide and completely prevented by NF-kappaB-binding site mutation in the mmp-9 promoter. Taken together, these results indicate that PKC-zeta plays a key role in the regulation of MMP-9 expression in C6 glioma cells through NF-kappaB.
Collapse
|
46
|
Santibáñez JF, Guerrero J, Quintanilla M, Fabra A, Martínez J. Transforming growth factor-beta1 modulates matrix metalloproteinase-9 production through the Ras/MAPK signaling pathway in transformed keratinocytes. Biochem Biophys Res Commun 2002; 296:267-73. [PMID: 12163012 DOI: 10.1016/s0006-291x(02)00864-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mouse transformed keratinocytes cultured in the presence of transforming growth factor-beta1 (TGF-beta1) acquire a set of morphological and functional properties giving rise to a more motile phenotype that expresses mesenchymal markers. In this work, we present evidence showing that TGF-beta1 stimulates cellular production of MMP-9 (Gelatinase B), a metalloproteinase that plays an important role in tumoral invasion. Our results demonstrate that TGF-beta1stimulates MMP-9 production and MMP-9 promoter activity in a process that depends of the activation of the Ras-ERK1,2 MAP kinase pathway. The latter was demonstrated by cellular transfection of TGF-beta1-sensitive cells with a RasN17 mutant gene, using PD 098059, a MEK 1,2 inhibitor, and treating cells with anti-sense oligodeoxinucleotides. The enhanced MMP-9 production proved to be an important factor in the acquisition of migratory and invasive properties as shown by the use of a specific inhibitor of MMP-9 (GM6001) that inhibits the TGF-beta1-stimulated invasive and migratory properties of these transformed keratinocytes.
Collapse
|
47
|
Lakka SS, Jasti SL, Gondi C, Boyd D, Chandrasekar N, Dinh DH, Olivero WC, Gujrati M, Rao JS. Downregulation of MMP-9 in ERK-mutated stable transfectants inhibits glioma invasion in vitro. Oncogene 2002; 21:5601-8. [PMID: 12165859 DOI: 10.1038/sj.onc.1205646] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2002] [Revised: 05/03/2002] [Accepted: 05/07/2002] [Indexed: 12/31/2022]
Abstract
We previously showed that enhanced expression of MMP-9, an endopeptidase that digests basement-membrane type IV collagen, is related to tumor progression in vitro and in vivo; antisense-MMP-9 stably transfected clones were less invasive than untransfected parental cells and did not form tumors in nude mice. In this study, we examined the role of ERK-1 in the regulation of MMP-9 production and the invasive behavior of the human glioblastoma cell line SNB19, in which ERK1 is constitutively activated. SNB19 cells were stably transfected with mt-ERK, a vector encoding ERK-1 cDNA in which the conserved lysine at codon 71 was changed to arginine, thus impairing the catalytic efficiency of this enzyme. Gelatin zymography showed reduced levels of MMP-9 in the mt-ERK-transfected cell lines relative to those in vector-transfected and parental control cells. Reductions in MMP-9 protein mRNA levels were also detected in the mt-ERK-transfected cells by Western and Northern blotting. The mt-ERK-transfected cells were much less invasive than parental or vector control cells in a Matrigel invasion assay and in a spheroid coculture assay. Thus an ERK-dependent signaling pathway seems to regulate MMP-9 mediated glioma invasion in SNB19 cells; interfering with this pathway could be developed into a therapeutic approach, which aims at a reduction of cancer cell invasion.
Collapse
Affiliation(s)
- Sajani S Lakka
- Division of Cancer Biology, Department of Biomedical and Therapeutic Sciences, University of Illinois College of Medicine at Peoria, 61656, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
VanMeter TE, Rooprai HK, Kibble MM, Fillmore HL, Broaddus WC, Pilkington GJ. The role of matrix metalloproteinase genes in glioma invasion: co-dependent and interactive proteolysis. J Neurooncol 2002. [PMID: 11716072 DOI: 10.1023/a: 1012280925031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Matrix metalloproteinases (MMPs) are cation-dependent endopeptidases which have been implicated in the malignancy of gliomas. It is thought that the MMPs play a critical role in both metastasis and angiogenesis, and that interference with proteases might therefore deter local tumor dissemination and neovascularization. However, the attempt to control tumor-associated proteolysis will rely on better definition of the normal tissue function of MMPs, an area of study still in its infancy in the central nervous system (CNS). Understanding the role of MMP-mediated proteolysis in the brain relies heavily on advances in other areas of molecular neuroscience, most notably an understanding of extracellular matrix (ECM) composition and the function of cell adhesion molecules such as integrins, which communicate knowledge of ECM composition intracellularly. Recently, protease expression and function has been shown to be strongly influenced by the functional state and signaling properties of integrins. Here we review MMP function and expression in gliomas and present examples of MMP profiling studies in glioma tissues and cell lines by RT-PCR and Western blotting. Co-expression of MMPs and certain integrins substantiates the gathering evidence of a functional intersection between the two, and inhibition studies using recombinant TIMP-1 and integrin antisera demonstrate significant inhibition of glioma invasion in vitro. Use of promising new therapeutic compounds with anti-MMP and anti-invasion effects are discussed. These data underline the importance of functional interaction of MMPs with accessory proteins such as integrins during invasion, and the need for further studies to elucidate the molecular underpinnings of this process.
Collapse
Affiliation(s)
- T E VanMeter
- Department of Neuropathology, Institute of Psychiatry, King's College London, De Crespigny Park, London, UK
| | | | | | | | | | | |
Collapse
|
49
|
Bauvois B, Dumont J, Mathiot C, Kolb JP. Production of matrix metalloproteinase-9 in early stage B-CLL: suppression by interferons. Leukemia 2002; 16:791-8. [PMID: 11986939 DOI: 10.1038/sj.leu.2402472] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2001] [Accepted: 01/14/2002] [Indexed: 11/09/2022]
Abstract
Besides vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), matrix metalloproteinases (MMPs) play critical roles in angiogenesis, tumor invasion and metastasis. Increased angiogenesis is observed in chronic B lymphocytic leukemia (B-CLL) and published data reported VEGF and bFGF production in this disease. The purpose of this study was to investigate MMP expression in early stage B-CLL. Elevated MMP-9 concentrations were detected by ELISA in the sera of B-CLL patients (median level 250 ng/ml) compared with healthy donors (67 ng/ml) (P < 0.0001), and immunostaining with antibodies against MMP-9 and B cell antigens (CD19, CD23) substantiated the presence of MMP-9 in tumoral B lymphocytes. By using RT-PCR, ELISA and zymography experiments, we confirmed that B-CLL cells expressed and released the pro-form of MMP-9 with Mr 92 kDa (158-1300 pg/ml/10(6) cells/48 h), p-aminophenylmercuric acetate generating a 82 kDa active form. In contrast, the production of MMP-9 by normal counterpart B cells was significantly low (28-169 pg/ml/10(6)cells/48 h). Moreover, B-CLL culture supernatants contained bFGF (median levels 17 pg/ml/10(6) cells/48 h), VEGF (1.4 pg/ml/10(6) cells/48 h) and TNF-alpha (0.2 pg/ml/10(6) cells/48 h). TNF-alpha and VEGF antibodies blocked MMP-9 at the mRNA and protein levels. Interferons (IFNs) type I or type II repressed MMP-9 gelatinolytic activity in a dose and time dependency, and this was reflected by a parallel inhibition of MMP-9 mRNA and protein. IFNs however did not affect the production of bFGF, VEGF and TNF-alpha. Together, our data show that B-CLL lymphocytes synthesize MMP-9 and emphasize the specific inhibitory actions of IFNs on its expression.
Collapse
Affiliation(s)
- B Bauvois
- Unité 365 INSERM, Section de Recherche, Institut Curie, Pavillon Pasteur, 26 rue d'Ulm, 75248 Paris cedex 05, France
| | | | | | | |
Collapse
|
50
|
Rivera-Marrero CA, Schuyler W, Roser S, Ritzenthaler JD, Newburn SA, Roman J. M. tuberculosis induction of matrix metalloproteinase-9: the role of mannose and receptor-mediated mechanisms. Am J Physiol Lung Cell Mol Physiol 2002; 282:L546-55. [PMID: 11839551 DOI: 10.1152/ajplung.00175.2001] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) infection induces the expression of matrix metalloproteinase-9 (MMP-9) in mouse lungs. In cultured human monocytic cells, Mtb bacilli and the cell wall glycolipid lipoarabinomannan (LAM) stimulate high levels of MMP-9 activity. Here, we explore the cellular mechanisms involved in the induction of MMP-9 by Mtb. We show that infection of THP-1 cells with Mtb caused a fivefold increase in MMP-9 mRNA that was associated with increased MMP-9 activity. MMP-9 induction was dependent on microtubule polymerization and protein kinase activation and was associated with increased DNA binding by the transcription factor activator protein-1 (AP-1), which appeared to be important for MMP-9 expression. We then explored the surface molecules potentially involved in Mtb induction of MMP-9, focusing on ligands of the mannose and beta-glucan receptors. MMP-9 activity was induced by the mannose receptor ligands mannan, zymosan, and LAM, whereas the beta-glucan receptor ligand laminarin was not effective. The most active inducers of MMP-9 activity were the particulate ligand zymosan and LAM. Pretreatment of cells with an anti-mannose receptor monoclonal antibody, but not anti-complement receptor 3, decreased the induction of MMP-9 activity by Mtb bacilli. Together, these results suggest that MMP-9 induction by Mtb occurs by receptor-mediated signaling mechanisms involving the binding of mannosylated ligands to mannose receptors, the modulation by cytoskeletal elements such as microtubules, the activation of protein kinases, and transcriptional activation by AP-1.
Collapse
Affiliation(s)
- Carlos A Rivera-Marrero
- Pulmonary and Critical Care Division, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University School of Medicine, Atlanta, Georgia 30033, USA
| | | | | | | | | | | |
Collapse
|