1
|
Bautista-Bautista G, Salguero-Zacarias S, Villeda-Gabriel G, García-López G, Osorio-Caballero M, Palafox-Vargas ML, Acuña-González RJ, Lara-Pereyra I, Díaz-Ruíz O, Flores-Herrera H. Escherichia coli induced matrix metalloproteinase-9 activity and type IV collagen degradation is regulated by progesterone in human maternal decidual. BMC Pregnancy Childbirth 2024; 24:645. [PMID: 39367340 PMCID: PMC11451097 DOI: 10.1186/s12884-024-06847-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/20/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Escherichia coli (E. coli) is one of the main bacteria associated with preterm premature rupture of membranes by increasing pro-matrix metalloproteinase 9 (proMMP-9) and degradation of type IV collagen in human feto-maternal interface (HFMi). proMMP-9 is regulated by progesterone (P4) but it is unclear whether P4 inhibits proMMP in human maternal decidual (MDec). This study aimed to determine a role of P4 on proMMP-2 and - 9 and type IV collagen induced by E. coli infection in MDec. METHODS Nine HFMi were mounted in a Transwell system. MDec was stimulated with P4 or E. coli for 3-, 6-, or 24-hours. proMMP-2, -9 and type IV collagen were assessed. RESULTS Gelatin zymography revealed an increase in proMMP-9 after 3, 6, and 24 h of stimulating MDec with E. coli. Using immunofluorescence, it was confirmed the increase in the HFMi tissue and a reduction on the amount of type IV collagen leading to the separation of fetal amniochorion and MDEc. The degradative activity of proMMP-9 was reduced by 20% by coincubation with P4. CONCLUSIONS P4 modulates the activity of proMMP-9 induced by E. coli stimulation but it was unable to completely reverse the degradation of type IV collagen in human MDec tissue.
Collapse
Affiliation(s)
- Gerardo Bautista-Bautista
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Urales #800 Col. Lomas de Virreyes CP 11000, Tercer piso de la Torre de Investigación, Ciudad de México, México
| | - Santos Salguero-Zacarias
- Departamento de Tococirugia y Urgencias, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, México
| | - Graciela Villeda-Gabriel
- Departamento de Inmunología e infectología, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, México
| | - Guadalupe García-López
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes , Ciudad de México, México
| | - Mauricio Osorio-Caballero
- Departamento de Salud Sexual y Reproductiva, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, México
| | - Martha Leticia Palafox-Vargas
- Departamento de Anatomía Patológica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, México
| | - Ricardo Josué Acuña-González
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Urales #800 Col. Lomas de Virreyes CP 11000, Tercer piso de la Torre de Investigación, Ciudad de México, México
| | - Irlando Lara-Pereyra
- Departamento de Ginecología, Hospital General de Zona 252, Instituto Mexicano del Seguro Social, Atlacomulco, México
| | - Oscar Díaz-Ruíz
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hector Flores-Herrera
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Urales #800 Col. Lomas de Virreyes CP 11000, Tercer piso de la Torre de Investigación, Ciudad de México, México.
| |
Collapse
|
2
|
Morales-Vázquez MM, Meza-Serrano E, Lara-Pereyra I, Acuña-González RJ, Alonso-Morales R, Hayen-Valles S, Boeta AM, Zarco L, Lozano-Cuenca J, López-Canales JS, Flores-Herrera H. Equine Placentitis in Mares Induces the Secretion of Pro-Inflammatory Cytokine eIL-1β and the Active Extracellular Matrix Metalloproteinase (MMP)-9. Vet Sci 2023; 10:532. [PMID: 37756054 PMCID: PMC10536981 DOI: 10.3390/vetsci10090532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/05/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
Equine placentitis is characterized by infection and inflammation of the placenta. Different biomarkers associated with this inflammatory response have been evaluated in experimentally induced equine placentitis, but not in pregnant mares with spontaneous placentitis. The aim of the current study was to determine the concentration of eIL-1β and the activity of proMMP-2 and proMMP-9 in the serum of healthy mares and mares with placentitis on days 240 and 320 of gestation to explore whether these biomarkers are associated with equine maternal placentitis and/or with the birth of an infected or inviable foals. Serum samples were collected from sixteen pregnant English Thoroughbred mares, retrospectively classified as follows: (1) healthy mares with full-term gestation; and (2) mares with ultrasonographic signs of placentitis. The health of each foal was examined at birth, and it was decided to classify the cases into four groups: (1) healthy mares delivering a healthy foals (HM-HF, n = 6); (2) mares with USP delivering a healthy foal (USP-HF, n = 3); (3) mares with USP delivering a live septic foal (USP-LSeF, n = 4); and (4) mares with USP delivering a dead foal (USP-DF, n = 3). eIL-1β was quantified by ELISA, and proMMP-2 and proMMP-9 activity by gelatin zymography electrophoresis. In healthy mares, the serum concentrations of eIL-1β underwent a significant 16.5-fold increase from day 240 to day 320 of gestation. Although similar results were found in the mares with ultrasonographic signs of placentitis that delivered a healthy foal, those delivering a live septic or nonviable foal exhibited much higher concentrations of eIL-1β. proMMP-2 and proMMP-9 activity was not associated with maternal placentitis, foal infection, or death. Hence, the presence of placentitis severe enough to affect the health of the foal can be confirmed or discarded by determining the eIL-1β concentration in mares that have shown ultrasonographic signs of placentitis.
Collapse
Affiliation(s)
- María Margarita Morales-Vázquez
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Europa Meza-Serrano
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Irlando Lara-Pereyra
- Departamento de Ginecología, Hospital General de Zona 252, Instituto Mexicano del Seguro Social, Atlacomulco 28984, Mexico
| | - Ricardo Josué Acuña-González
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
| | - Rogelio Alonso-Morales
- Genética, Laboratorio de Biotecnologías, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico;
| | - Sergio Hayen-Valles
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Ana Myriam Boeta
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Luis Zarco
- Centro de Enseñanza, Investigación y Extensión en Producción Ovina, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Tres Marías, Ciudad de México 62515, Mexico;
| | - Jair Lozano-Cuenca
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, Mexico; (J.L.-C.); (J.S.L.-C.)
| | - Jorge Skiold López-Canales
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, Mexico; (J.L.-C.); (J.S.L.-C.)
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Héctor Flores-Herrera
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
| |
Collapse
|
3
|
Tanabe H, Suzuki T, Ohishi T, Isemura M, Nakamura Y, Unno K. Effects of Epigallocatechin-3-Gallate on Matrix Metalloproteinases in Terms of Its Anticancer Activity. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020525. [PMID: 36677584 PMCID: PMC9862901 DOI: 10.3390/molecules28020525] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023]
Abstract
Epidemiological studies have shown that the consumption of green tea has beneficial effects against cancer. Basic studies have provided evidence that epigallocatechin gallate (EGCG) is a major contributor to these effects. Matrix metalloproteinases (MMPs) are zinc-dependent metalloproteinases with the ability to degrade the extracellular matrix proteins and are involved in various diseases including cancer in which MMPs have a critical role in invasion and metastasis. In this review, we discuss the effects of EGCG on several types of MMPs in the context of its anticancer activity. In the promoter region, MMPs have binding sites for at least one transcription factor of AP-1, Sp1, and NF-κB, and EGCG can downregulate these transcription factors through signaling pathways mediated by reactive oxygen species. EGCG can also decrease nuclear ERK, p38, heat shock protein-27 (Hsp27), and β-catenin levels, leading to suppression of MMPs' expression. Other mechanisms by which EGCG inhibits MMPs include direct binding to MMPs to prevent their activation and downregulation of NF-κB to suppress the production of inflammatory cytokines such as TNFα and IL-1β. Findings from studies on EGCG presented here may be useful in the development of more effective anti-MMP agents, which would give beneficial effects on cancer and other diseases.
Collapse
Affiliation(s)
- Hiroki Tanabe
- Faculty of Health and Welfare Science, Nayoro City University, Nayoro 096-8641, Hokkaido, Japan
- Correspondence: (H.T.); (T.O.)
| | - Takuji Suzuki
- Department of Food Science and Nutrition, Faculty of Human Life and Science, Doshisha Women’s College of Liberal Arts, Kyoto 602-0893, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu 410-0301, Shizuoka, Japan
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, Shinagawa, Tokyo 141-0021, Japan
- Correspondence: (H.T.); (T.O.)
| | - Mamoru Isemura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yoriyuki Nakamura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Keiko Unno
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
4
|
Wang J, Zhang N, Peng M, Hua X, Huang C, Tian Z, Xie Q, Zhu J, Li J, Huang H, Huang C. p85α Inactivates MMP-2 and Suppresses Bladder Cancer Invasion by Inhibiting MMP-14 Transcription and TIMP-2 Degradation. Neoplasia 2019; 21:908-920. [PMID: 31401412 PMCID: PMC6700442 DOI: 10.1016/j.neo.2019.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023]
Abstract
Recent studies show p85α up-regulates epidermal growth factor (EGF) receptor, thereby promoting malignant cell transformation and migration in normal mouse embryonic fibroblasts (MEFs). However, the potential role of p85α in human bladder cancer (BC) remains unknown. Here, we show that p85α is down-regulated in BC tumor tissues. Ectopic expression of p85α inhibited cell invasion, but not migration, whereas p85α knockdown promoted invasion in BC cells, revealing that p85α inhibits BC invasion. Overexpression of kinase-deficient p110 in T24 T(p85α) cells inhibited BC cell migration, but not invasion, suggesting that the inhibition of p85α on invasion is independent of PI3K activity. The effect of p85α on inhibiting BC invasion was mediated by the inactivation of MMP-2 concomitant with the up-regulation of TIMP-2 and down-regulation of MMP-14. Mechanistic studies revealed c-Jun inactivation was associated with p85α knockdown-induced MMP-14 expression, and down-regulated miR-190, leading to ATG7 mRNA degradation. This suppressed the autophagy-dependent removal of TIMP-2 in human BC cells. The present results identify a novel function of p85α and clarify the mechanisms underlying its inhibition of BC invasion, providing insight into the role of p85α in normal and cancer cells.
Collapse
Affiliation(s)
- Jingjing Wang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Ning Zhang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Minggang Peng
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10010, USA
| | - Xiaohui Hua
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10010, USA
| | - Chao Huang
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10010, USA
| | - Zhongxian Tian
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10010, USA
| | - Qipeng Xie
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Junlan Zhu
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10010, USA
| | - Jingxia Li
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10010, USA
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035.
| | - Chuanshu Huang
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10010, USA.
| |
Collapse
|
5
|
Chang J, Chaudhuri O. Beyond proteases: Basement membrane mechanics and cancer invasion. J Cell Biol 2019; 218:2456-2469. [PMID: 31315943 PMCID: PMC6683740 DOI: 10.1083/jcb.201903066] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022] Open
Abstract
In epithelial cancers, cells must invade through basement membranes (BMs) to metastasize. The BM, a thin layer of extracellular matrix underlying epithelial and endothelial tissues, is primarily composed of laminin and collagen IV and serves as a structural barrier to cancer cell invasion, intravasation, and extravasation. BM invasion has been thought to require protease degradation since cells, which are typically on the order of 10 µm in size, are too large to squeeze through the nanometer-scale pores of the BM. However, recent studies point toward a more complex picture, with physical forces generated by cancer cells facilitating protease-independent BM invasion. Moreover, collective cell interactions, proliferation, cancer-associated fibroblasts, myoepithelial cells, and immune cells are all implicated in regulating BM invasion through physical forces. A comprehensive understanding of BM structure and mechanics and diverse modes of BM invasion may yield new strategies for blocking cancer progression and metastasis.
Collapse
Affiliation(s)
- Julie Chang
- Department of Bioengineering, Stanford University, Stanford, CA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA
| |
Collapse
|
6
|
Chalcone Derivatives 4'-Amino-1-Naphthyl-Chalcone (D14) and 4'-Amino-4-Methyl-1-Naphthyl-Chalcone (D15) Suppress Migration and Invasion of Osteosarcoma Cells Mediated by p53 Regulating EMT-Related Genes. Int J Mol Sci 2018; 19:ijms19092838. [PMID: 30235848 PMCID: PMC6163733 DOI: 10.3390/ijms19092838] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/04/2018] [Accepted: 09/04/2018] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor that mainly affects children, adolescents, and young adults. The inhibition of metastasis is a main strategy of OS therapy since the development of metastatic disease due to drug resistance remains the most important cause of death from this cancer. Considering the severe side effects of current OS chemotherapy, the identification of anti-metastatic drugs with reduced toxicity is of great interest. Chalcones are polyphenols with a basic structure consisting of an α-, β-unsaturated carbonyl system linking two aryl rings. These compounds exhibit anticancer activity against a variety of tumor cell lines through multiple mechanisms, including the regulation of the tumor-suppressor protein p53 and its target genes. An important process regulated by p53 is epithelial-mesenchymal transition (EMT), which facilitates tumor metastasis by conferring migratory and invasive properties to cancer cells. The activation of p53 can revert EMT and reduce migration and invasion. This study aimed to examine the inhibitory effects of two 4′-aminochalcones on the migration/invasion of the U2OS (p53+/+) and SAOS-2 (p53−/−) OS cell lines as well as the underlying molecular mechanisms. Cell viability was examined by MTT assay. Transwell assays were used to evaluate the migratory and invasive ability of the cells. The two 4′-aminochalcones showed low capacity to inhibit the viability of OS cells independent of p53 status, but preferentially suppressed the migration of U2OS cells and of a SAOS-2 cell line expressing p53. Invasion was strongly inhibited by both chalcones independent of p53 status. RT-PCR, zymography, and Western blot were used to study the expression of matrix metalloproteinases and EMT markers after treatment with the chalcones. The results indicated that the 4′-aminochalcone-induced antimigratory and anti-invasive effects are potentially associated with the inhibition of extracellular matrix (ECM) enzymatic degradation in OS cells and with the modulation of EMT genes. These effects probably result from the induced increase of p53 protein expression by the two chalcones. In conclusion, chalcones D14 and D15 have potential anti-metastatic activity mediated by p53 that can be exploited for OS treatment.
Collapse
|
7
|
Silva G, Teixeira Lima F, Seba V, Mendes Lourenço AL, Lucas TG, de Andrade BV, Torrezan GS, Polaquini CR, Garcia ME, Couto LB, Bestetti RB, de Castro França S, Fachin AL, Regasini LO, Marins M. Curcumin Analog CH-5 Suppresses the Proliferation, Migration, and Invasion of the Human Gastric Cancer Cell Line HGC-27. Molecules 2018; 23:E279. [PMID: 29385675 PMCID: PMC6017500 DOI: 10.3390/molecules23020279] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the most frequent malignant tumors in the world. The majority of patients are diagnosed with metastatic gastric cancer, which has a low survival rate. These data reinforce the importance of studying the anticancer activity of new molecules with the potential to suppress gastric cancer metastasis. Curcumin is a well-studied compound that has demonstrated anti-metastatic effects. Here we investigated if CH-5, a curcumin derivative compound, has anti-metastatic properties in the human gastric cancer cell line HGC-27. Firstly, we found that CH-5 decreased viability and induced apoptosis in HGC-27 cells in a dose-dependent manner. Additionally, CH-5 suppressed the migration and invasion of HGC-27 cells by downregulating the expression and collagenase activity of matrix metalloproteinase 2 in a dose-dependent manner. In conclusion, CH-5 showed anticancer activities, including the induction of apoptosis, and the suppression of migration and invasion in HGC-27 cells, suggesting that CH-5 can be a lead molecule for the development of anti-metastatic drugs for gastric cancer therapy.
Collapse
Affiliation(s)
- Gabriel Silva
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
| | - Felipe Teixeira Lima
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
| | - Viviane Seba
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
| | - Ana Laura Mendes Lourenço
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (M.E.G.); (L.B.C.); (R.B.B.)
| | - Thaise Graminha Lucas
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (M.E.G.); (L.B.C.); (R.B.B.)
| | - Bianca Vieira de Andrade
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (M.E.G.); (L.B.C.); (R.B.B.)
| | - Guilherme Silva Torrezan
- Laboratory of Green and Medicinal Chemistry, Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, SP CEP 15054-000, Brazil; (G.S.T.); (C.R.P.)
| | - Carlos Roberto Polaquini
- Laboratory of Green and Medicinal Chemistry, Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, SP CEP 15054-000, Brazil; (G.S.T.); (C.R.P.)
| | - Marcelo Engracia Garcia
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (M.E.G.); (L.B.C.); (R.B.B.)
| | - Lucélio Bernardes Couto
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (M.E.G.); (L.B.C.); (R.B.B.)
| | - Reinaldo Bulgarelli Bestetti
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (M.E.G.); (L.B.C.); (R.B.B.)
| | - Suzelei de Castro França
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
| | - Ana Lúcia Fachin
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (M.E.G.); (L.B.C.); (R.B.B.)
| | - Luis Octavio Regasini
- Laboratory of Green and Medicinal Chemistry, Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, SP CEP 15054-000, Brazil; (G.S.T.); (C.R.P.)
| | - Mozart Marins
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (G.S.); (F.T.L.); (V.S.); (A.L.M.L.); (T.G.L.); (B.V.d.A.); (S.d.C.F.); (A.L.F.)
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, SP CEP 14096-900, Brazil; (M.E.G.); (L.B.C.); (R.B.B.)
| |
Collapse
|
8
|
Abstract
There is currently no effective treatment for multiorgan failure following shock other than supportive care. A better understanding of the pathogenesis of these sequelae to shock is required. The intestine plays a central role in multiorgan failure. It was previously suggested that bacteria and their toxins are responsible for the organ failure seen in circulatory shock, but clinical trials in septic patients have not confirmed this hypothesis. Instead, we review here evidence that the digestive enzymes, synthesized in the pancreas and discharged into the small intestine as requirement for normal digestion, may play a role in multiorgan failure. These powerful enzymes are nonspecific, highly concentrated, and fully activated in the lumen of the intestine. During normal digestion they are compartmentalized in the lumen of the intestine by the mucosal epithelial barrier. However, if this barrier becomes permeable, e.g. in an ischemic state, the digestive enzymes escape into the wall of the intestine. They digest tissues in the mucosa and generate small molecular weight cytotoxic fragments such as unbound free fatty acids. Digestive enzymes may also escape into the systemic circulation and activate other degrading proteases. These proteases have the ability to clip the ectodomain of surface receptors and compromise their function, for example cleaving the insulin receptor causing insulin resistance. The combination of digestive enzymes and cytotoxic fragments leaking into the central circulation causes cell and organ dysfunction, and ultimately may lead to complete organ failure and death. We summarize current evidence suggesting that enteral blockade of digestive enzymes inside the lumen of the intestine may serve to reduce acute cell and organ damage and improve survival in experimental shock.
Collapse
|
9
|
Horejs CM. Basement membrane fragments in the context of the epithelial-to-mesenchymal transition. Eur J Cell Biol 2016; 95:427-440. [PMID: 27397693 DOI: 10.1016/j.ejcb.2016.06.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 01/18/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) enables cells of epithelial phenotype to become motile and change to a migratory mesenchymal phenotype. EMT is known to be a fundamental requisite for tissue morphogenesis, and EMT-related pathways have been described in cancer metastasis and tissue fibrosis. Epithelial structures are marked by the presence of a sheet-like extracellular matrix, the basement membrane, which is assembled from two major proteins, laminin and collagen type IV. This specialized matrix is essential for tissue function and integrity, and provides an important barrier to the potential pathogenic migration of cells. The profound phenotypic transition in EMT involves the epithelial cells disrupting the basement membrane. Matrix metalloproteinases (MMPs) are known to cleave components of basement membranes, but MMP-basement membrane crosstalk during EMT in vivo is poorly understood. However, MMPs have been reported to play a role in EMT-related processes and a variety of basement membrane fragments have been shown to be released by specific MMPs in vitro and in vivo exhibiting distinct biological activities. This review discusses general considerations regarding the basement membrane in the context of EMT, a possible role for specific MMPs in EMT and highlights biologically active basement membrane fragments liberated by MMPs.
Collapse
Affiliation(s)
- Christine-Maria Horejs
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles vaeg 2, 17177 Stockholm, Sweden.
| |
Collapse
|
10
|
Kim Y, Williams KC, Gavin CT, Jardine E, Chambers AF, Leong HS. Quantification of cancer cell extravasation in vivo. Nat Protoc 2016; 11:937-48. [PMID: 27101515 DOI: 10.1038/nprot.2016.050] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer cell 'invasiveness' is one of the main driving forces in cancer metastasis, and assays that quantify this key attribute of cancer cells are crucial in cancer metastasis research. The research goal of many laboratories is to elucidate the signaling pathways and effectors that are responsible for cancer cell invasion, but many of these experiments rely on in vitro methods that do not specifically simulate individual steps of the metastatic cascade. Cancer cell extravasation is arguably the most important example of invasion in the metastatic cascade, whereby a single cancer cell undergoes transendothelial migration, forming invasive processes known as invadopodia to mediate translocation of the tumor cell from the vessel lumen into tissue in vivo. We have developed a rapid, reproducible and economical technique to evaluate cancer cell invasiveness by quantifying in vivo rates of cancer cell extravasation in the chorioallantoic membrane (CAM) of chicken embryos. This technique enables the investigator to perform well-powered loss-of-function studies of cancer cell extravasation within 24 h, and it can be used to identify and validate drugs with potential antimetastatic effects that specifically target cancer cell extravasation. A key advantage of this technique over similar assays is that intravascular cancer cells within the capillary bed of the CAM are clearly distinct from extravasated cells, which makes cancer cell extravasation easy to detect. An intermediate level of experience in injections of the chorioallantoic membrane of avian embryos and cell culture techniques is required to carry out the protocol.
Collapse
Affiliation(s)
- Yohan Kim
- Department of Surgery, Schulich School of Medicine, Western University, London, Ontario, Canada.,Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, Ontario, Canada
| | - Karla C Williams
- Department of Surgery, Schulich School of Medicine, Western University, London, Ontario, Canada.,Translational Breast Cancer Research Unit, London Health Sciences Centre, London, Ontario, Canada
| | - Carson T Gavin
- Department of Surgery, Schulich School of Medicine, Western University, London, Ontario, Canada.,Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, Ontario, Canada
| | - Emily Jardine
- Department of Surgery, Schulich School of Medicine, Western University, London, Ontario, Canada.,Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, Ontario, Canada
| | - Ann F Chambers
- Translational Breast Cancer Research Unit, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, Schulich School of Medicine, Western University, London, Ontario, Canada
| | - Hon S Leong
- Department of Surgery, Schulich School of Medicine, Western University, London, Ontario, Canada.,Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
11
|
Gasparics Á, Rosivall L, Krizbai IA, Sebe A. When the endothelium scores an own goal: endothelial cells actively augment metastatic extravasation through endothelial-mesenchymal transition. Am J Physiol Heart Circ Physiol 2016; 310:H1055-63. [PMID: 26993222 DOI: 10.1152/ajpheart.00042.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/14/2016] [Indexed: 01/05/2023]
Abstract
Endothelial-mesenchymal transition (EndMT) is an important mechanism during organ development and in certain pathological conditions. For example, EndMT contributes to myofibroblast formation during organ fibrosis, and it has been identified as an important source of cancer-associated fibroblasts, facilitating tumor progression. Recently, EndMT was proposed to modulate endothelial function during intravasation and extravasation of metastatic tumor cells. Evidence suggests that endothelial cells are not passive actors during transendothelial migration (TEM) of cancer cells, as there are profound changes in endothelial junctional protein expression, signaling, permeability, and contractility. This review describes these alterations in endothelial characteristics during TEM of metastatic tumor cells and discusses them in the context of EndMT. EndMT could play an important role during metastatic intravasation and extravasation, a novel hypothesis that may lead to new therapeutic approaches to tackle metastatic disease.
Collapse
Affiliation(s)
- Ákos Gasparics
- Department of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - László Rosivall
- Department of Pathophysiology, Semmelweis University, Budapest, Hungary; Pediatrics and Nephrology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - István A Krizbai
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary; Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania; and
| | - Attila Sebe
- Department of Pathophysiology, Semmelweis University, Budapest, Hungary; Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| |
Collapse
|
12
|
Klein D, Schmetter A, Imsak R, Wirsdörfer F, Unger K, Jastrow H, Stuschke M, Jendrossek V. Therapy with Multipotent Mesenchymal Stromal Cells Protects Lungs from Radiation-Induced Injury and Reduces the Risk of Lung Metastasis. Antioxid Redox Signal 2016; 24:53-69. [PMID: 26066676 DOI: 10.1089/ars.2014.6183] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIMS Previous thorax irradiation promotes metastatic spread of tumor cells to the lung. We hypothesized that vascular damage facilitates lung metastasis after thorax irradiation and that therapeutically applied multipotent mesenchymal stromal cells (MSCs) with reported repair activity may prevent these adverse effects of ionizing radiation by protecting lung endothelia from radiation-induced damage. RESULTS Previous whole-thorax irradiation (WTI) with 15 Gy significantly enhanced seeding and metastatic growth of tumor cells in the lung. WTI was further associated with endothelial cell damage, senescence of lung epithelial cells, and upregulation of invasion- and inflammation-promoting soluble factors, for example, endothelial matrix metalloproteinase 2 (Mmp2), its activator Mmp14, the cofactor tissue inhibitor of metalloproteinases 2 (Timp2), chemokine (C-C motif) ligand 2 (Ccl2), and urokinase-type plasminogen activator (Plau/uPA), and recruitment of CD11b+CD11c- myelomonocytic cells. Inhibition of Mmp2 counteracted radiation-induced vascular dysfunction without preventing increased metastasis. In contrast, therapy with bone marrow or aorta-derived MSCs within 2 weeks postirradiation antagonized radiation-induced damage to resident cells as well as the resulting secretome changes and abrogated the metastasis-promoting effects of WTI. INNOVATION Therapy with MSCs protects lungs from radiation-induced injury and reduces the risk of lung metastasis. MSC-mediated inhibition of Mmp2 mediates their protective effects at the vasculature. Furthermore, local and systemic effects such as inhibition of radiation-induced senescence of bronchial epithelial cells and associated secretion of immunomodulatory factors may participate in the inhibitory effect of MSCs on lung metastasis. CONCLUSION MSC therapy is a promising strategy to prevent radiation-induced lung injury and the resulting increased risk of metastasis.
Collapse
Affiliation(s)
- Diana Klein
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Alexandra Schmetter
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Roze Imsak
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Florian Wirsdörfer
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Kristian Unger
- 2 Research Unit Radiation Cytogenetics, Helmholtz-Zentrum München, German Research Center for Environmental Health , Neuherberg, Germany and Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer', Helmholtz-Zentrum München, Neuherberg, Germany
| | - Holger Jastrow
- 3 Institute of Anatomy, University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Martin Stuschke
- 4 Department of Radiotherapy, University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Verena Jendrossek
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| |
Collapse
|
13
|
Correlation of Forkhead Box c2 with subtypes and invasive ability of invasive breast cancer. ACTA ACUST UNITED AC 2014; 34:896-901. [DOI: 10.1007/s11596-014-1370-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 08/31/2014] [Indexed: 10/24/2022]
|
14
|
Afsharimani B, Cabot P, Parat MO. Effect of lysine antifibrinolytics and cyclooxygenase inhibitors on the proteolytic profile of breast cancer cells interacting with macrophages or endothelial cells. Br J Anaesth 2014; 113 Suppl 1:i22-31. [DOI: 10.1093/bja/aet468] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
15
|
Voura EB, English JL, Yu HYE, Ho AT, Subarsky P, Hill RP, Hojilla CV, Khokha R. Proteolysis during tumor cell extravasation in vitro: metalloproteinase involvement across tumor cell types. PLoS One 2013; 8:e78413. [PMID: 24194929 PMCID: PMC3806793 DOI: 10.1371/journal.pone.0078413] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/20/2013] [Indexed: 12/22/2022] Open
Abstract
To test if proteolysis is involved in tumor cell extravasation, we developed an in vitro model where tumor cells cross an endothelial monolayer cultured on a basement membrane. Using this model we classified the ability of the cells to transmigrate through the endothelial cell barrier onto the underlying matrix, and scored this invasion according to the stage of passage through the endothelium. Metalloproteinase inhibitors reduced tumor cell extravasation by at least 35%. Visualization of protease and cell adhesion molecules by confocal microscopy demonstrated the cell surface localization of MMP-2, MMP-9, MT1-MMP, furin, CD44 and αvβ3, during the process of transendothelial migration. By the addition of inhibitors and bio-modulators we assessed the functional requirement of the aforementioned molecules for efficient migration. Proteolytic digestion occurred at the cell-matrix interface and was most evident during the migratory stage. All of the inhibitors and biomodulators affected the transition of the tumor cells into the migratory stage, highlighting the most prevalent use of proteolysis at this particular step of tumor cell extravasation. These data suggest that a proteolytic interface operates at the tumor cell surface within the tumor-endothelial cell microenvironment.
Collapse
Affiliation(s)
- Evelyn B. Voura
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
- Department of Biology, Dominican College, Orangeburg, New York, United States of America
| | - Jane L. English
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Hoi-Ying E. Yu
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Andrew T. Ho
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Patrick Subarsky
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Richard P. Hill
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Carlo V. Hojilla
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Rama Khokha
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
16
|
Afsharimani B, Baran J, Watanabe S, Lindner D, Cabot PJ, Parat MO. Morphine and breast tumor metastasis: the role of matrix-degrading enzymes. Clin Exp Metastasis 2013; 31:149-58. [DOI: 10.1007/s10585-013-9616-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/01/2013] [Indexed: 12/16/2022]
|
17
|
Panduratin A, a possible inhibitor in metastasized A549 cells through inhibition of NF-kappa B translocation and chemoinvasion. Molecules 2013; 18:8764-78. [PMID: 23887718 PMCID: PMC6270481 DOI: 10.3390/molecules18088764] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/21/2013] [Accepted: 06/28/2013] [Indexed: 01/01/2023] Open
Abstract
In the present study, we investigated the effects of panduratin A (PA), isolated from Boesenbergia rotunda, on apoptosis and chemoinvasion in A549 human non-small cell lung cancer cells. Activation of the executioner procaspase-3 by PA was found to be dose-dependent. Caspase-3 activity was significantly elevated at the 5 µg/mL level of PA treatment and progressed to a maximal level. However, no significant elevated level was detected on procaspase-8. These findings suggest that PA activated caspase-3 but not caspase-8. Numerous nuclei of PA treated A549 cells stained brightly by anti-cleaved PARP antibody through High Content Screening. This result further confirmed that PA induced apoptotic cell death was mediated through activation of caspase-3 and eventually led to PARP cleavage. Treatment of A549 cells with PA resulted in a strong inhibition of NF-κB activation, which was consistent with a decrease in nuclear levels of NF-κB/p65 and NF-κB/p50 and the elevation of p53 and p21. Besides that, we also showed that PA significantly inhibited the invasion of A549 cells in a dose-dependent manner through reducing the secretion of MMP-2 of A549 cells gelatin zymography assay. Our findings not only provide the effects of PA, but may also be important in the design of therapeutic protocols that involve targeting of either p53 or NF-κB.
Collapse
|
18
|
Aref AR, Huang RYJ, Yu W, Chua KN, Sun W, Tu TY, Bai J, Sim WJ, Zervantonakis IK, Thiery JP, Kamm RD. Screening therapeutic EMT blocking agents in a three-dimensional microenvironment. Integr Biol (Camb) 2013; 5:381-9. [PMID: 23172153 DOI: 10.1039/c2ib20209c] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Epithelial-mesenchymal transition (EMT) plays a critical role in the early stages of dissemination of carcinoma leading to metastatic tumors, which are responsible for over 90% of all cancer-related deaths. Current therapeutic regimens, however, have been ineffective in the cure of metastatic cancer, thus an urgent need exists to revisit existing protocols and to improve the efficacy of newly developed therapeutics. Strategies based on preventing EMT could potentially contribute to improving the outcome of advanced stage cancers. To achieve this goal new assays are needed to identify targeted drugs capable of interfering with EMT or to revert the mesenchymal-like phenotype of carcinoma to an epithelial-like state. Current assays are limited to examining the dispersion of carcinoma cells in isolation in conventional 2-dimensional (2D) microwell systems, an approach that fails to account for the 3-dimensional (3D) environment of the tumor or the essential interactions that occur with other nearby cell types in the tumor microenvironment. Here we present a microfluidic system that integrates tumor cell spheroids in a 3D hydrogel scaffold, in close co-culture with an endothelial monolayer. Drug candidates inhibiting receptor activation or signal transduction pathways implicated in EMT have been tested using dispersion of A549 lung adenocarcinoma cell spheroids as a metric of effectiveness. We demonstrate significant differences in response to drugs between 2D and 3D, and between monoculture and co-culture.
Collapse
Affiliation(s)
- Amir R Aref
- BioSystems and Micromechanics IRG, S16-07, SMART, Singapore 117543, Singapore
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
LI FENGSHENG, GAO LING, WANG ZHIDONG, DONG BO, YAN TAO, JIANG QISHENG, CHEN XIAOHUA. Radiation enhances the invasion abilities of pulmonary adenocarcinoma cells via STAT3. Mol Med Rep 2013; 7:1883-8. [DOI: 10.3892/mmr.2013.1441] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 04/12/2013] [Indexed: 11/06/2022] Open
|
20
|
Lee JH, Shim JW, Choi YJ, Heo K, Yang K. The combination of sorafenib and radiation preferentially inhibits breast cancer stem cells by suppressing HIF-1α expression. Oncol Rep 2013; 29:917-24. [PMID: 23314174 PMCID: PMC3597559 DOI: 10.3892/or.2013.2228] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 12/10/2012] [Indexed: 01/27/2023] Open
Abstract
The importance of anticancer stem cell research for breast cancer lies in the possibility of providing new approaches for an improved understanding of anticancer activity and cancer treatment. In this study, we demonstrated that the preclinical therapeutic efficacy of combining the multikinase inhibitor sorafenib with radiation was more effective in hypoxia-exposed breast cancer stem cells. We assessed cell viability and Annexin V to evaluate the combined effect of sorafenib and radiation following exposure to hypoxia. Our results showed that the synergistic cytotoxicity increased tumor cell apoptosis significantly and reduced cell proliferation in MDA-MB-231 and MCF-7 cells under hypoxic conditions compared to sorafenib or radiation alone in vitro. Additionally, the combined treatment induced G2/M cell cycle arrest. Notably, the combination of sorafenib and radiation eliminated CD44+CD24-/low cells preferentially, which highly expressed hypoxia-inducible factor (HIF)-1α and effectively inhibited primary and secondary mammosphere formation in MDA-MB-231 cells. A combined effect on MDA-MB‑231 cells in response to hypoxia was shown by inhibiting angiogenesis and metastasis by suppression of HIF-1α and matrix metalloproteinase-2 (MMP-2). Collectively, these results indicate that the efficacy of sorafenib combined with radiation for treating human breast cancer cells is synergistic and suggest a new therapeutic approach to prevent breast cancer progression by eliminating breast cancer stem cells.
Collapse
Affiliation(s)
- Jae Ho Lee
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, Republic of Korea
| | | | | | | | | |
Collapse
|
21
|
Katunina AI, Gershtein ES, Ermilova VD, Tereshkina IV, Nazarenko AY, Tyleuova AA, Dvorova EK, Karabekova ZK, Gritskevich MV, Berezov TT. Matrix metalloproteinases 2, 7, and 9 in tumors and sera of patients with breast cancer. Bull Exp Biol Med 2012; 151:359-62. [PMID: 22451887 DOI: 10.1007/s10517-011-1330-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Enzyme immunoassay showed that the content of matrix metalloproteinases (MMP) 2 and 7 in tumors was higher than in the adjacent histologically intact tissue in 91 and 76% patients with breast cancer, respectively, while MMP-9 levels in the tumor and intact tissue were virtually the same. Serum concentrations of MMP-2 and MMP-7 did not correlate with their levels in the tumors, were within the normal range, and virtually did not decrease after removal of the primary tumor. Serum levels of MMP-9 in patients were significantly lower than in the control and increased after surgery in 85% patients. No clear-cut relationship between the studied parameters and clinical morphological prognostic factors of breast cancer was detected.
Collapse
Affiliation(s)
- A I Katunina
- N. N. Blokhin Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lai SL, Cheah SC, Wong PF, Noor SM, Mustafa MR. In vitro and in vivo anti-angiogenic activities of Panduratin A. PLoS One 2012; 7:e38103. [PMID: 22666456 PMCID: PMC3364190 DOI: 10.1371/journal.pone.0038103] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Targeting angiogenesis has emerged as an attractive and promising strategy in anti-cancer therapeutic development. The present study investigates the anti-angiogenic potential of Panduratin A (PA), a natural chalcone isolated from Boesenbergia rotunda by using both in vitro and in vivo assays. METHODOLOGY/PRINCIPAL FINDINGS PA exerted selective cytotoxicity on human umbilical vein endothelial cells (HUVECs) with IC(50) value of 6.91 ± 0.85 µM when compared to human normal fibroblast and normal liver epithelial cells. Assessment of the growth kinetics by cell impedance-based Real-Time Cell Analyzer showed that PA induced both cytotoxic and cytostatic effects on HUVECs, depending on the concentration used. Results also showed that PA suppressed VEGF-induced survival and proliferation of HUVECs. Furthermore, endothelial cell migration, invasion, and morphogenesis or tube formation demonstrated significant time- and dose-dependent inhibition by PA. PA also suppressed matrix metalloproteinase-2 (MMP-2) secretion and attenuated its activation to intermediate and active MMP-2. In addition, PA suppressed F-actin stress fiber formation to prevent migration of the endothelial cells. More importantly, anti-angiogenic potential of PA was also evidenced in two in vivo models. PA inhibited neo-vessels formation in murine Matrigel plugs, and angiogenesis in zebrafish embryos. CONCLUSIONS/SIGNIFICANCE Taken together, our study demonstrated the distinctive anti-angiogenic properties of PA, both in vitro and in vivo. This report thus reveals another biological activity of PA in addition to its reported anti-inflammatory and anti-cancer activities, suggestive of PA's potential for development as an anti-angiogenic agent for cancer therapy.
Collapse
Affiliation(s)
- Siew-Li Lai
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Suzita Mohd Noor
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohd Rais Mustafa
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Lee ES, Shen Q, Pitts RL, Guo M, Wu MH, Sun SC, Yuan SY. Serum metalloproteinases MMP-2, MMP-9, and metalloproteinase tissue inhibitors in patients are associated with arteriovenous fistula maturation. J Vasc Surg 2011; 54:454-9; discussion 459-60. [PMID: 21620625 PMCID: PMC3171276 DOI: 10.1016/j.jvs.2011.02.056] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2010] [Revised: 02/22/2011] [Accepted: 02/22/2011] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Many vascular surgeons construct arteriovenous fistulas (AVFs) for hemodialysis access as the primary choice access. A significant number of AVFs fail to mature, however, leading to patient frustration and repeated operations. Metalloproteinase (MMP) activity, particularly MMP-2 and MMP-9, may be important for AVF maturation. We therefore sought to identify whether serum MMP levels could serve as a biomarker for predicting future successful AVF maturation. METHODS Blood was collected from patients with chronic renal insufficiency at the time of surgery for long-term hemodialysis access. Serum was separated from whole blood and ultracentrifuged at 1000g for 10 minutes. Serum aliquots were frozen at -80°C until used for analysis. Enzyme-linked immunosorbent assay was used to assay levels of MMP-2, MMP-9, and tissue inhibitor of metalloproteinase type 2 (TIMP-2), and TIMP type 4 (TIMP-4). Clinical end points were used to divide patients into failed and matured AVF groups. Successful maturation was considered in patients who had specific duplex findings or 1 month of successful two-needle cannulation hemodialysis. MMP/TIMP ratios were calculated as an index of the MMP axis activity because MMP activity parallels alterations in TIMP levels. RESULTS Of 20 enrolled patients, AVF maturation was successful in 13 and failed in 7. Serum levels of MMP-2/TIMP-2 were significantly higher in patients with matured AVFs vs levels in those that failed (P = .003). Similarly, a trend toward increased serum levels of MMP-9/TIMP-4 was found in patients with successful AVF (P = .06). CONCLUSIONS MMP-2 and TIMP-2 levels were different among patients whose AVF matured vs those who did not. Further follow-up studies to determine the predictability of AVF maturation using relative patient serum levels of MMP-2 and TIMP-2 should be performed.
Collapse
Affiliation(s)
- Eugene S Lee
- Department of Surgery, University of California, Davis, Sacramento, CA, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Calvo F, Sahai E. Cell communication networks in cancer invasion. Curr Opin Cell Biol 2011; 23:621-9. [PMID: 21570276 DOI: 10.1016/j.ceb.2011.04.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 04/14/2011] [Accepted: 04/18/2011] [Indexed: 01/07/2023]
Abstract
The invasion of cancer is a major clinical problem. It is now apparent that invasion is not a simply a cancer cell autonomous process but relies on a complex network of paracrine interactions. Furthermore, this network can change as cancer cells disseminate. Here we summarise the key components of the network and their mechanisms of communication. Finally, we discuss the difficulties and opportunities that this complex network of interactions presents during cancer therapy.
Collapse
Affiliation(s)
- Fernando Calvo
- Tumour Cell Biology Laboratory, London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, UK
| | | |
Collapse
|
25
|
Yue P, Gao ZH, Xue X, Cui SX, Zhao CR, Yuan Y, Yin Z, Inagaki Y, Kokudo N, Tang W, Qu XJ. Des-γ-carboxyl prothrombin induces matrix metalloproteinase activity in hepatocellular carcinoma cells by involving the ERK1/2 MAPK signalling pathway. Eur J Cancer 2011; 47:1115-24. [PMID: 21349701 DOI: 10.1016/j.ejca.2011.01.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 01/18/2011] [Accepted: 01/26/2011] [Indexed: 12/22/2022]
Abstract
Des-γ-carboxy prothrombin (DCP), an aberrant prothrombin produced by hepatocellular carcinoma (HCC) cells, has been shown to be associated with the biological malignant potential of HCC. The aim of this study was to evaluate the effect of DCP on HCC cell growth and metastasis, and to explore the underlying molecular mechanisms. DCP significantly stimulated HCC cell growth, as measured by cell counting kit-8 assay. Transwell chamber assay showed that DCP increased HCC cell migration through reconstituted extracellular matrix (Matrigel). Gelatin zymography assay and Western blot analysis demonstrated that DCP increased the secretion and expression of matrix metalloproteinase (MMP)-2 and MMP-9 in the supernatant of cultured HCC cells and on tumour cell membranes. DCP was found to bind to the cell surface receptor Met, resulting in Met phosphorylation and subsequent activation of the epidermal growth factor receptor (EGFR). Western blot analysis demonstrated that DCP stimulated a sequential kinase phosphorylation cascade including ERK1/2, MEK1/2 and c-Raf, indicating activation of the extracellular signal-regulated kinase/mitogen activated protein kinase (ERK1/2 MAPK) signalling pathway. Furthermore, blocking ERK1/2 MAPK activation with ERK1/2 inhibitor PD98059 essentially abolished the DCP-induced MMP-2 and MMP-9 activity, confirming the signalling pathway of DCP stimulation. Taken together, these results suggested that DCP stimulates HCC growth and promotes HCC metastasis by increasing the activity of MMP-2 and MMP-9 through activation of the ERK1/2 MAPK signalling pathway.
Collapse
Affiliation(s)
- Pan Yue
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Lee ES, Shen Q, Pitts RL, Guo M, Wu MH, Yuan SY. Vein tissue expression of matrix metalloproteinase as biomarker for hemodialysis arteriovenous fistula maturation. Vasc Endovascular Surg 2010; 44:674-9. [PMID: 20724289 PMCID: PMC5584062 DOI: 10.1177/1538574410377021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Failure of arteriovenous fistula (AVF) maturation is attributed to impaired vein remodeling. The purpose of this study is to identify whether vein matrix metalloproteinase (MMP) expression and activity is associated with AVF maturation. Patients with renal insufficiency undergoing surgery had their vein segments harvested and snap-frozen at time of AVF construction. Expression of MMP-2, MMP-9, membrane type-1 MMP (MT1-MMP), tissue inhibitor of metallopreoteinases type 2 (TIMP-2), and TIMP-4 were measured using zymography and Western blotting techniques. Of 14 patients enrolled, 9 had successful maturation and 5 had failure of AVF maturation. Significantly higher levels of MT1-MMP (an MMP-2 activator; P = .01), TIMP-2 (an MMP-2 inhibitor; P = .03), MMP-2 latent (P = .02), and MMP-2 total (P = .03) were associated with AVF maturation. There was a trend toward higher levels of TIMP-4 in the successful group (P = .18). These data demonstrate a positive relationship between MMP-2 expression in veins and AVF maturation. MMP-2 could serve as a potential preoperative marker to predict maturation.
Collapse
Affiliation(s)
- Eugene S Lee
- Department of Surgery, University of California, Davis, CA, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Shen Q, Lee ES, Pitts RL, Wu MH, Yuan SY. Tissue inhibitor of metalloproteinase-2 regulates matrix metalloproteinase-2-mediated endothelial barrier dysfunction and breast cancer cell transmigration through lung microvascular endothelial cells. Mol Cancer Res 2010; 8:939-51. [PMID: 20571065 PMCID: PMC5584073 DOI: 10.1158/1541-7786.mcr-09-0523] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Matrix metalloproteinases (MMP) have been implicated in multiple stages of cancer metastasis. Tissue inhibitor of metalloproteinase-2 (TIMP-2) plays an important role in regulating MMP-2 activity. By forming a ternary complex with pro-MMP-2 and its activator MMP-14 on the cell surface, TIMP-2 can either initiate or restrain the cleavage and subsequent activation of MMP-2. Our recent work has shown that breast cancer cell adhesion to vascular endothelial cells activates endothelial MMP-2, promoting tumor cell transendothelial migration (TEM(E)). However, the mechanism of MMP-2 regulation during TEM(E) remains unclear. In the current study, we present evidence that MMP-14 is expressed in both invasive breast cancer cells (MDA-MB-231 and MDA-MB-436) and lung microvascular endothelial cells (HBMVEC-L), whereas TIMP-2 is exclusively expressed and released from the cancer cells. The tumor cell-derived TIMP-2 was further identified as a major determinant of endothelial MMP-2 activity during tumor cell transmigration in the presence of MMP-14. This response was associated with endothelial barrier dysfunction because coculture of MDA-MB-231 or MDA-MB-436 with HBMVEC-L caused a significant decrease in transendothelial electrical resistance concomitantly with endothelial cell-cell junction disruption and tumor cell transmigration. Knockdown of TIMP-2 or inhibition of TIMP-2/MMP-14 attenuated MMP-2-dependent transendothelial electrical resistance response and TEM(E). These findings suggest a novel interactive role of breast cancer cells and vascular endothelial cells in regulating the TIMP-2/MMP-14/MMP-2 pathway during tumor metastasis.
Collapse
Affiliation(s)
- Qiang Shen
- Division of Research, Department of Surgery, University of California at Davis School of Medicine, Sacramento, California 95817, USA
| | | | | | | | | |
Collapse
|
29
|
van der Heul-Nieuwenhuijsen L, Dits N, Van Ijcken W, de Lange D, Jenster G. The FOXF2 pathway in the human prostate stroma. Prostate 2009; 69:1538-47. [PMID: 19562724 DOI: 10.1002/pros.20996] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Forkhead box 2 (FOXF2) is a member of the large family of forkhead transcription factors and its expression pattern suggests a role in prostate cancer development. FOXF2 expression is stroma-specific and higher expressed in the prostate transition zone than the prostate peripheral zone. Moreover, expression of FOXF2 is decreased in prostate cancer. METHODS To identify the genes and pathways regulated by FOXF2, we compared microarray expression profiles of primary prostate stromal cells (PrSC) treated with control or small interfering RNA (siRNA) directed against FOXF2. RESULTS From our microarray analyses, we selected 190 differentially expressed genes, of which 104 genes were higher expressed in PrSC cells treated with FOXF2 siRNA and 86 were higher expressed in PrSC cells treated with negative control siRNA. Eight of the strongest differentially expressed genes were validated by RT-PCR. Genes down-regulated by FOXF2 included MT1E, MT1F, PDGFA, ITGB1, and PSG7 and genes up-regulated by FOXF2 included WASF2, BAMBI, and CXCL12. Ingenuity pathway analysis showed several pathways significantly regulated by FOXF2, including PPAR signaling, PDGF signaling, and extracellular matrix (ECM) signaling. GSEA analysis revealed that FOXF2 up-regulated genes were down-regulated in the same PrSC cells treated with transforming growth factor 3 (TGFbeta3). CONCLUSIONS The distinct expression pattern of FOXF2 in the prostate, its effect on expression of ECM signaling, and its opposing role in the TGFbeta3 pathway, suggests a role for FOXF2 in prostate homeostasis and stroma-epithelial interactions.
Collapse
|
30
|
Molecular mechanisms of endothelial hyperpermeability: implications in inflammation. Expert Rev Mol Med 2009; 11:e19. [PMID: 19563700 DOI: 10.1017/s1462399409001112] [Citation(s) in RCA: 281] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endothelial hyperpermeability is a significant problem in vascular inflammation associated with trauma, ischaemia-reperfusion injury, sepsis, adult respiratory distress syndrome, diabetes, thrombosis and cancer. An important mechanism underlying this process is increased paracellular leakage of plasma fluid and protein. Inflammatory stimuli such as histamine, thrombin, vascular endothelial growth factor and activated neutrophils can cause dissociation of cell-cell junctions between endothelial cells as well as cytoskeleton contraction, leading to a widened intercellular space that facilitates transendothelial flux. Such structural changes initiate with agonist-receptor binding, followed by activation of intracellular signalling molecules including calcium, protein kinase C, tyrosine kinases, myosin light chain kinase, and small Rho-GTPases; these kinases and GTPases then phosphorylate or alter the conformation of different subcellular components that control cell-cell adhesion, resulting in paracellular hypermeability. Targeting key signalling molecules that mediate endothelial-junction-cytoskeleton dissociation demonstrates a therapeutic potential to improve vascular barrier function during inflammatory injury.
Collapse
|
31
|
Kang SW, Choi JS, Bae JY, Li J, Kim DS, Kim JL, Shin SY, You HJ, Park HS, Ji GE, Kang YH. Blockade of vascular angiogenesis by Aspergillus usamii var. shirousamii-transformed Angelicae Gigantis Radix and Zizyphus jujuba. Nutr Res Pract 2009; 3:3-8. [PMID: 20016695 PMCID: PMC2788165 DOI: 10.4162/nrp.2009.3.1.3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 01/22/2009] [Accepted: 02/03/2009] [Indexed: 12/03/2022] Open
Abstract
The matrix metalloproteinases (MMP) play an important role in tumor invasion, angiogenesis and inflammatory tissue destruction. Increased expression of MMP was observed in benign tissue hyperplasia and in atherosclerotic lesions. Invasive cancer cells utilize MMP to degrade the extracellular matrix and vascular basement membrane during metastasis, where MMP-2 has been implicated in the development and dissemination of malignancies. The present study attempted to examine the antiangiogenic activity of the medicinal herbs of Aspergillus usamii var. shirousamii-transformed Angelicae Gigantis Radix and Zizyphus jujube (tAgR and tZj) with respect to MMP-2 production and endothelial motility in phorbol 12-myristate 13-acetate (PMA)- or VEGF-exposed human umbilical vein endothelial cells (HUVEC). Nontoxic tAgR and tZj substantially suppressed PMA-induced MMP-2 secretion. In addition, 25 µg/mL tAgR and tZj prevented vascular endothelial growth factor-stimulated endothelial cell transmigration and tube formation. The results reveal that tAgR and tZj dampened endothelial MMP-2 production leading to endothelial transmigration and tube formation. tAgR and tZj-mediated inhibition of endothelial MMP may boost a therapeutic efficacy during vascular angiogenesis.
Collapse
Affiliation(s)
- Sang-Wook Kang
- Department of Food and Nutrition and Korean Institute of Nutrition, Hallym University, 39 Hallymdaehak-gil, Chuncheon, Kangwon 200-702, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Des-gamma-carboxy prothrombin stimulates human vascular endothelial cell growth and migration. Clin Exp Metastasis 2009; 26:469-77. [PMID: 19263229 DOI: 10.1007/s10585-009-9246-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 02/23/2009] [Indexed: 02/07/2023]
Abstract
Des-gamma-carboxy prothrombin (DCP) is an aberrant prothrombin produced by hepatocellular carcinoma (HCC) cells. Serum and tissue DCP expressions are thought to reflect the biological malignant potential of HCC. However, the role of DCP in the development of angiogenesis is not well understood. Herein, we report the effects of DCP on growth and migration of human vascular endothelial cells. DCP significantly stimulated the proliferation of HUVEC (ECV304) cells in a dose and time dependent manner, as measured by the MTT assay. A continuous rapid migration of ECV304 cells was observed in the presence of DCP measured by the scratch wound assay. The continuous rapid invasive activity, measured by transwell chamber assay also showed that DCP increased endothelial cells migration through the reconstituted extracellular matrix (Matrigel). Further, the tube formation of vascular endothelial cells on 3-D Matrigel showed an increased number of branch points of ECV304 cells induced by DCP in a dose dependent manner. The levels of vascular endothelial cell growth-related angiogenic factors and matrix metalloproteinase were also examined. DCP significantly stimulated the expression levels of epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), and matrix metalloproteinase (MMP)-2 (latent and active). Together, these data suggest that DCP is a novel type of vascular endothelial growth factor that possesses potent mitogenic and migrative activities in angiogenesis of HCC.
Collapse
|