1
|
Huang Q, Wang J, Ning H, Liu W, Han X. Integrin β1 in breast cancer: mechanisms of progression and therapy. Breast Cancer 2025; 32:43-59. [PMID: 39343856 DOI: 10.1007/s12282-024-01635-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
The therapy for breast cancer (BC), to date, still needs improvement. Apart from traditional therapy methods, biological therapy being explored opens up a novel avenue for BC patients. Integrin β1 (ITGβ1), one of the largest subgroups in integrin family, is a key player in cancer evolution and therapy. Recent researches progress in the relationship of ITGβ1 level and BC, finding that ITGβ1 expression evidently concerns BC progression. In this chapter, we outline diverse ITGβ1-based mechanisms regarding to the promoted effect of ITGβ1 on BC cell structure rearrangement and malignant phenotype behaviors, the unfavorable patient prognosis conferred by ITGβ1, BC therapy tolerance induced by ITGβ1, and lastly novel inhibitors targeting ITGβ1 for BC therapy. As an effective biomarker, ITGβ1 undoubtedly emerges one of targeted-therapy opportunities of BC patients in future. It is a necessity focusing on scientific and large-scale clinical trials on the validation of targeted-ITGβ1 drugs for BC patients.
Collapse
Affiliation(s)
- Qionglian Huang
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jue Wang
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanjuan Ning
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Liu
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianghui Han
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
2
|
Cerutti C, Lucotti S, Menendez ST, Reymond N, Garg R, Romero IA, Muschel R, Ridley AJ. IQGAP1 and NWASP promote human cancer cell dissemination and metastasis by regulating β1-integrin via FAK and MRTF/SRF. Cell Rep 2024; 43:113989. [PMID: 38536816 DOI: 10.1016/j.celrep.2024.113989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/01/2024] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
Attachment of circulating tumor cells to the endothelial cells (ECs) lining blood vessels is a critical step in cancer metastatic colonization, which leads to metastatic outgrowth. Breast and prostate cancers are common malignancies in women and men, respectively. Here, we observe that β1-integrin is required for human prostate and breast cancer cell adhesion to ECs under shear-stress conditions in vitro and to lung blood vessel ECs in vivo. We identify IQGAP1 and neural Wiskott-Aldrich syndrome protein (NWASP) as regulators of β1-integrin transcription and protein expression in prostate and breast cancer cells. IQGAP1 and NWASP depletion in cancer cells decreases adhesion to ECs in vitro and retention in the lung vasculature and metastatic lung nodule formation in vivo. Mechanistically, NWASP and IQGAP1 act downstream of Cdc42 to increase β1-integrin expression both via extracellular signal-regulated kinase (ERK)/focal adhesion kinase signaling at the protein level and by myocardin-related transcription factor/serum response factor (SRF) transcriptionally. Our results identify IQGAP1 and NWASP as potential therapeutic targets to reduce early metastatic dissemination.
Collapse
Affiliation(s)
- Camilla Cerutti
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 U1L, UK; Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge UB8 3PH, UK.
| | - Serena Lucotti
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Sofia T Menendez
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 U1L, UK
| | - Nicolas Reymond
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 U1L, UK
| | - Ritu Garg
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 U1L, UK
| | - Ignacio A Romero
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes MK7 6AA, UK
| | - Ruth Muschel
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Anne J Ridley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 U1L, UK.
| |
Collapse
|
3
|
Mutgan AC, Jandl K, Radic N, Valzano F, Kolb D, Hoffmann J, Foris V, Wilhelm J, Boehm PM, Hoetzenecker K, Olschewski A, Olschewski H, Heinemann A, Wygrecka M, Marsh LM, Kwapiszewska G. Pentastatin, a matrikine of the collagen IVα5, is a novel endogenous mediator of pulmonary endothelial dysfunction. Am J Physiol Cell Physiol 2023; 325:C1294-C1312. [PMID: 37694286 PMCID: PMC11550886 DOI: 10.1152/ajpcell.00391.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
Deposition of basement membrane components, such as collagen IVα5, is associated with altered endothelial cell function in pulmonary hypertension. Collagen IVα5 harbors a functionally active fragment within its C-terminal noncollageneous (NC1) domain, called pentastatin, whose role in pulmonary endothelial cell behavior remains unknown. Here, we demonstrate that pentastatin serves as a mediator of pulmonary endothelial cell dysfunction, contributing to pulmonary hypertension. In vitro, treatment with pentastatin induced transcription of immediate early genes and proinflammatory cytokines and led to a functional loss of endothelial barrier integrity in pulmonary arterial endothelial cells. Mechanistically, pentastatin leads to β1-integrin subunit clustering and Rho/ROCK activation. Blockage of the β1-integrin subunit or the Rho/ROCK pathway partially attenuated the pentastatin-induced endothelial barrier disruption. Although pentastatin reduced the viability of endothelial cells, smooth muscle cell proliferation was induced. These effects on the pulmonary vascular cells were recapitulated ex vivo in the isolated-perfused lung model, where treatment with pentastatin-induced swelling of the endothelium accompanied by occasional endothelial cell apoptosis. This was reflected by increased vascular permeability and elevated pulmonary arterial pressure induced by pentastatin. This study identifies pentastatin as a mediator of endothelial cell dysfunction, which thus might contribute to the pathogenesis of pulmonary vascular disorders such as pulmonary hypertension.NEW & NOTEWORTHY This study is the first to show that pentastatin, the matrikine of the basement membrane (BM) collagen IVα5 polypeptide, triggers rapid pulmonary arterial endothelial cell barrier disruption, activation, and apoptosis in vitro and ex vivo. Mechanistically, pentastatin partially acts through binding to the β1-integrin subunit and the Rho/ROCK pathway. These findings are the first to link pentastatin to pulmonary endothelial dysfunction and, thus, suggest a major role for BM-matrikines in pulmonary vascular diseases such as pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Nemanja Radic
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Francesco Valzano
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Dagmar Kolb
- Core Facility Ultrastructure Analysis, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Julia Hoffmann
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Vasile Foris
- Division of Pulmonology, Medical University of Graz, Graz, Austria
| | - Jochen Wilhelm
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| | - Panja M Boehm
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pulmonology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Malgorzata Wygrecka
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
- Center for Infection and Genomics of the Lung, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Leigh M Marsh
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Grazyna Kwapiszewska
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| |
Collapse
|
4
|
Dou R, Liu K, Yang C, Zheng J, Shi D, Lin X, Wei C, Zhang C, Fang Y, Huang S, Song J, Wang S, Xiong B. EMT-cancer cells-derived exosomal miR-27b-3p promotes circulating tumour cells-mediated metastasis by modulating vascular permeability in colorectal cancer. Clin Transl Med 2021; 11:e595. [PMID: 34936736 PMCID: PMC8694332 DOI: 10.1002/ctm2.595] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/04/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Metastasis is the main cause of death in colorectal cancer (CRC). Circulating tumour cells (CTCs) are regarded as the precursor cells of metastasis. The CTCs, which underwent epithelial-mesenchymal transition (EMT), are associated with metastasis and responsible for poor prognosis. EMT cancer cells modulate endothelial permeability in the invasive front and facilitate cancer cell intravasation, resulting in CTCs-mediated distant metastasis. Exosomes derived from cancer cells are key mediators of cancer-host intercommunication. However, the mechanism by which EMT-tumour cells-derived exosomes modulate vascular permeability and promote CTCs generation has remained unclear. METHODS Exosomes isolation and purification were conducted by ultra-centrifugation. Exosomal miRNA was identified by sequencing followed by quantitative PCR. In vitro co-culture assay experiments were conducted to evaluate the effect of exosomal miR-27b-3p on the permeability of blood vessel endothelium. Dual-luciferase reporter assay, chromatin immunoprecipitation (ChIP) and RNA immunoprecipitation (RIP) were performed to investigate the underlying mechanism by which miR-27b-3p is packaged into exosomes. A mouse model was established to determine the role of exosomal miR-27b-3p in blood vessel permeability modulation in vivo. RESULTS We found that EMT-CRC cells attenuate the blood vessel barrier by transferring miR-27b-3p to human umbilical vein endothelial cells (HUVECs) in exosomes. Mechanically, miR-27b-3p atteuated the expression of vascular endothelial cadherin (VE-Cad) and p120 at the post-transcriptional level by binding to 3'-untranslated region of VE-Cad and p120 directly. The packaging of miR-27b-3p into exosomes was induced by heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), which activated by STAT3. Clinically, miR-27b-3p up-regulated in CRC tissues. Plasma exosomal miR-27b-3p was positively correlated with malignant progression and CTC count in CRC patients. Our study reveals a novel mechanism by which EMT-CRC cells promote metastasis, increasing blood vessel permeability and facilitating the generation of CTCs. CONCLUSION Exosomal miR-27b-3p secreted by EMT-CRC cells increases blood vessel permeability and facilitates the generation of CTCs. Exosomal miR-27b-3p may become a promising biomarker for CRC metastasis.
Collapse
|
5
|
Rodenburg WS, van Buul JD. Rho GTPase signalling networks in cancer cell transendothelial migration. VASCULAR BIOLOGY 2021; 3:R77-R95. [PMID: 34738075 PMCID: PMC8558887 DOI: 10.1530/vb-21-0008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/21/2023]
Abstract
Rho GTPases are small signalling G-proteins that are central regulators of cytoskeleton dynamics, and thereby regulate many cellular processes, including the shape, adhesion and migration of cells. As such, Rho GTPases are also essential for the invasive behaviour of cancer cells, and thus involved in several steps of the metastatic cascade, including the extravasation of cancer cells. Extravasation, the process by which cancer cells leave the circulation by transmigrating through the endothelium that lines capillary walls, is an essential step for metastasis towards distant organs. During extravasation, Rho GTPase signalling networks not only regulate the transmigration of cancer cells but also regulate the interactions between cancer and endothelial cells and are involved in the disruption of the endothelial barrier function, ultimately allowing cancer cells to extravasate into the underlying tissue and potentially form metastases. Thus, targeting Rho GTPase signalling networks in cancer may be an effective approach to inhibit extravasation and metastasis. In this review, the complex process of cancer cell extravasation will be discussed in detail. Additionally, the roles and regulation of Rho GTPase signalling networks during cancer cell extravasation will be discussed, both from a cancer cell and endothelial cell point of view.
Collapse
Affiliation(s)
- Wessel S Rodenburg
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands.,Leeuwenhoek Centre for Advanced Microscopy, Section Molecular Cytology at Swammerdam Institute for Life Sciences at University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
6
|
Zhang Z, Xiong X, Zhang R, Xiong G, Yu C, Xu L. Bioinformatics analysis reveals biomarkers with cancer stem cell characteristics in kidney renal clear cell carcinoma. Transl Androl Urol 2021; 10:3501-3514. [PMID: 34532274 PMCID: PMC8421844 DOI: 10.21037/tau-21-647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/16/2021] [Indexed: 11/06/2022] Open
Abstract
Background Kidney renal clear cell carcinoma (KIRC) is a renal cortical tumor. KIRC is the most common subtype of kidney cancer, accounting for 70%-80% of kidney cancer. Early identification of the risk of KIRC patients can facilitate more accurate clinical treatment, but there is a lack of effective prognostic markers. We aimed to identify new prognostic biomarkers for KIRC on the basis of the cancer stem cell (CSC) theory. Methods RNA-sequencing (RNA-seq) data and related clinical information were downloaded from The Cancer Genome Atlas (TCGA) database. Weighted gene co-expression network analysis (WGCNA) was used to identify significant modules and hub genes, and predictive hub genes were used to construct prognostic characteristics. Results The messenger RNA expression-based stemness index (mRNAsi) in tumor tissues of patients in the TCGA database is higher than that of the corresponding normal tissues. In addition, some clinical features and results are highly correlated with mRNAsi. WGCNA found that the green module is the most prominent module associated with mRNAsi; the genes in the green module are mainly concentration in Notch binding, endothelial cell development, Notch signaling pathway, and Rap 1 signaling pathway. A protein-protein interaction (PPI) network showed that the top 10 central genes were significantly associated with the transcriptional level. Moreover, the 10 hub genes were up-regulated in KIRC. Regarding survival analysis, the nomogram of the prognostic markers of the seven pivotal genes showed a higher predictive value. The classical receiver operating characteristic (ROC) curve analysis showed that risk score biomarkers had the highest accuracy and specificity with an area under the curve (AUC) value of 0.701. Conclusions mRNAsi-related genes may be good prognostic biomarkers for KIRC.
Collapse
Affiliation(s)
- Zan Zhang
- College of Life Sciences and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xueyang Xiong
- College of Life Sciences and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Rufeng Zhang
- College of Life Sciences and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Guoliang Xiong
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Changyuan Yu
- College of Life Sciences and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lida Xu
- College of Life Sciences and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
7
|
Sigdel I, Gupta N, Faizee F, Khare VM, Tiwari AK, Tang Y. Biomimetic Microfluidic Platforms for the Assessment of Breast Cancer Metastasis. Front Bioeng Biotechnol 2021; 9:633671. [PMID: 33777909 PMCID: PMC7992012 DOI: 10.3389/fbioe.2021.633671] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 12/27/2022] Open
Abstract
Of around half a million women dying of breast cancer each year, more than 90% die due to metastasis. Models necessary to understand the metastatic process, particularly breast cancer cell extravasation and colonization, are currently limited and urgently needed to develop therapeutic interventions necessary to prevent breast cancer metastasis. Microfluidic approaches aim to reconstitute functional units of organs that cannot be modeled easily in traditional cell culture or animal studies by reproducing vascular networks and parenchyma on a chip in a three-dimensional, physiologically relevant in vitro system. In recent years, microfluidics models utilizing innovative biomaterials and micro-engineering technologies have shown great potential in our effort of mechanistic understanding of the breast cancer metastasis cascade by providing 3D constructs that can mimic in vivo cellular microenvironment and the ability to visualize and monitor cellular interactions in real-time. In this review, we will provide readers with a detailed discussion on the application of the most up-to-date, state-of-the-art microfluidics-based breast cancer models, with a special focus on their application in the engineering approaches to recapitulate the metastasis process, including invasion, intravasation, extravasation, breast cancer metastasis organotropism, and metastasis niche formation.
Collapse
Affiliation(s)
- Indira Sigdel
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Niraj Gupta
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Fairuz Faizee
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Vishwa M Khare
- Eurofins Lancaster Laboratories, Philadelphia, PA, United States
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Yuan Tang
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| |
Collapse
|
8
|
Stojak M, Milczarek M, Kurpinska A, Suraj-Prazmowska J, Kaczara P, Wojnar-Lason K, Banach J, Stachowicz-Suhs M, Rossowska J, Kalviņš I, Wietrzyk J, Chlopicki S. Protein Disulphide Isomerase A1 Is Involved in the Regulation of Breast Cancer Cell Adhesion and Transmigration via Lung Microvascular Endothelial Cells. Cancers (Basel) 2020; 12:cancers12102850. [PMID: 33023153 PMCID: PMC7601413 DOI: 10.3390/cancers12102850] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer cell cross-talk with the host endothelium plays a crucial role in metastasis, but the underlying mechanisms are still not fully understood. We studied the involvement of protein disulphide isomerase A1 (PDIA1) in human breast cancer cell (MCF-7 and MDA-MB-231) adhesion and transendothelial migration. For comparison, the role of PDIA1 in proliferation, migration, cell cycle and apoptosis was also assessed. Pharmacological inhibitor, bepristat 2a and PDIA1 silencing were used to inhibit PDIA1. Inhibition of PDIA1 by bepristat 2a markedly decreased the adhesion of breast cancer cells to collagen type I, fibronectin and human lung microvascular endothelial cells. Transendothelial migration of breast cancer cells across the endothelial monolayer was also inhibited by bepristat 2a, an effect not associated with changes in ICAM-1 expression or changes in cellular bioenergetics. The silencing of PDIA1 produced less pronounced anti-adhesive effects. However, inhibiting extracellular free thiols by non-penetrating blocker p-chloromercuribenzene sulphonate substantially inhibited adhesion. Using a proteomic approach, we identified that β1 and α2 integrins were the most abundant among all integrins in breast cancer cells as well as in lung microvascular endothelial cells, suggesting that integrins could represent a target for PDIA1. In conclusion, extracellular PDIA1 plays a major role in regulating the adhesion of cancer cells and their transendothelial migration, in addition to regulating cell cycle and caspase 3/7 activation by intracellular PDIA1. PDIA1-dependent regulation of cancer-endothelial cell interactions involves disulphide exchange and most likely integrin activation but is not mediated by the regulation of ICAM-1 expression or changes in cellular bioenergetics in breast cancer or endothelial cells.
Collapse
Affiliation(s)
- Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
| | - Magdalena Milczarek
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
| | - Anna Kurpinska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
| | - Joanna Suraj-Prazmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
| | - Patrycja Kaczara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
| | - Kamila Wojnar-Lason
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland
| | - Joanna Banach
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
| | - Martyna Stachowicz-Suhs
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
| | - Joanna Rossowska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
| | - Ivars Kalviņš
- Laboratory of Carbofunctional Compounds, Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia;
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
- Correspondence: (J.W.); (S.C.)
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland
- Correspondence: (J.W.); (S.C.)
| |
Collapse
|
9
|
Gadde M, Phillips C, Ghousifam N, Sorace AG, Wong E, Krishnamurthy S, Syed A, Rahal O, Yankeelov TE, Woodward WA, Rylander MN. In vitro vascularized tumor platform for modeling tumor-vasculature interactions of inflammatory breast cancer. Biotechnol Bioeng 2020; 117:3572-3590. [PMID: 32648934 DOI: 10.1002/bit.27487] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/24/2020] [Accepted: 07/08/2020] [Indexed: 12/26/2022]
Abstract
Inflammatory breast cancer (IBC), a rare form of breast cancer associated with increased angiogenesis and metastasis, is largely driven by tumor-stromal interactions with the vasculature and the extracellular matrix (ECM). However, there is currently a lack of understanding of the role these interactions play in initiation and progression of the disease. In this study, we developed the first three-dimensional, in vitro, vascularized, microfluidic IBC platform to quantify the spatial and temporal dynamics of tumor-vasculature and tumor-ECM interactions specific to IBC. Platforms consisting of collagen type 1 ECM with an endothelialized blood vessel were cultured with IBC cells, MDA-IBC3 (HER2+) or SUM149 (triple negative), and for comparison to non-IBC cells, MDA-MB-231 (triple negative). Acellular collagen platforms with endothelialized blood vessels served as controls. SUM149 and MDA-MB-231 platforms exhibited a significantly (p < .05) higher vessel permeability and decreased endothelial coverage of the vessel lumen compared to the control. Both IBC platforms, MDA-IBC3 and SUM149, expressed higher levels of vascular endothelial growth factor (p < .05) and increased collagen ECM porosity compared to non-IBCMDA-MB-231 (p < .05) and control (p < .01) platforms. Additionally, unique to the MDA-IBC3 platform, we observed progressive sprouting of the endothelium over time resulting in viable vessels with lumen. The newly sprouted vessels encircled clusters of MDA-IBC3 cells replicating a key feature of in vivo IBC. The IBC in vitro vascularized platforms introduced in this study model well-described in vivo and clinical IBC phenotypes and provide an adaptable, high throughput tool for systematically and quantitatively investigating tumor-stromal mechanisms and dynamics of tumor progression.
Collapse
Affiliation(s)
- Manasa Gadde
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Caleb Phillips
- Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas
| | - Neda Ghousifam
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas
| | - Anna G Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama.,Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama.,O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Enoch Wong
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Anum Syed
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Omar Rahal
- M.D. Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas.,Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas.,Departments of Diagnostic Medicine, The University of Texas at Austin, Austin, Texas.,Department of Oncology, The University of Texas at Austin, Austin, Texas.,Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Wendy A Woodward
- M.D. Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Marissa N Rylander
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas.,Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas.,Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
10
|
Shao Y, Zhong P, Sheng L, Zheng H. Circular RNA circDENND2A protects H9c2 cells from oxygen glucose deprivation-induced apoptosis through sponging microRNA-34a. Cell Cycle 2019; 19:246-255. [PMID: 31878833 DOI: 10.1080/15384101.2019.1708029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background/Aims: Myocardial ischemia (MI) is a serious threat to human health. Circular RNAs (circRNAs) play an important role in many diseases including MI. The effect and mechanism of circDENND2A in MI have not been studied.Methods: We used oxygen glucose deprivation (OGD) treatment to simulate MI in vitro. We detected circDENND2A and microRNA (miR)-34a levels by RT-qPCR. The transfection process used INTERFER and jetPRIME. Cell growth indexes including viability, apoptosis, and migration were detected by CCK8, flow cytometry, and transwell assays, respectively. In addition, the Bax, Cleaved-Caspase-3, matrix metalloproteinase (MMP)-2, MMP-9 and pathway-related protein levels were tested by Western blot.Results: OGD upregulated circDENND2A expression in H9c2 cells. Overexpression of circDENND2A enhanced cell viability and migration but declined apoptosis under OGD. Silenced circDENND 2A played the opposite effects. circDENND2A negatively regulated miR-34a. miR-34a overexpression weakened the protective effects of circDENND2A in OGD-injury. Moreover, we considered circDENND2A and miR-34a may work via β-catenin and Ras/Raf/MEK/ERK pathways.Conclusion: circDENND2A overexpression enhanced OGD-inhibited cell viability and migration but declined OGD-promoted apoptosis by downregulating miR-34a and via β-catenin and Ras/Raf/MEK/ERK pathways.
Collapse
Affiliation(s)
- Yuanxia Shao
- Department of Cardiology, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Peng Zhong
- Department of Cardiology, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Li Sheng
- Department of Cardiology, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Hongjian Zheng
- Department of Cardiology, Jining No.1 People's Hospital, Jining, Shandong, China
| |
Collapse
|
11
|
Lefley D, Howard F, Arshad F, Bradbury S, Brown H, Tulotta C, Eyre R, Alférez D, Wilkinson JM, Holen I, Clarke RB, Ottewell P. Development of clinically relevant in vivo metastasis models using human bone discs and breast cancer patient-derived xenografts. Breast Cancer Res 2019; 21:130. [PMID: 31783893 PMCID: PMC6884811 DOI: 10.1186/s13058-019-1220-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/25/2019] [Indexed: 12/29/2022] Open
Abstract
Background Late-stage breast cancer preferentially metastasises to bone; despite advances in targeted therapies, this condition remains incurable. The lack of clinically relevant models for studying breast cancer metastasis to a human bone microenvironment has stunted the development of effective treatments for this condition. To address this problem, we have developed humanised mouse models in which breast cancer patient-derived xenografts (PDXs) metastasise to human bone implants with low variability and high frequency. Methods To model the human bone environment, bone discs from femoral heads of patients undergoing hip replacement surgery were implanted subcutaneously into NOD/SCID mice. For metastasis studies, 7 patient-derived xenograft tumours (PDX: BB3RC32, ER+ PR+ HER2−; BB2RC08, ER+ PR+ ER2−; BB6RC37, ER− PR− HER2− and BB6RC39, ER+ PR+ HER2+), MDA-MB-231-luc2, T47D-luc2 or MCF7-Luc2 cells were injected into the 4th mammary ducts and metastases monitored by luciferase imaging and confirmed on histological sections. Bone integrity, viability and vascularisation were assessed by uCT, calcein uptake and histomorphometry. Expression profiling of genes/proteins during different stages of metastasis were assessed by whole genome Affymetrix array, real-time PCR and immunohistochemistry. Importance of IL-1 was confirmed following anakinra treatment. Results Implantation of femoral bone provided a metabolically active, human-specific site for tumour cells to metastasise to. After 4 weeks, bone implants were re-vascularised and demonstrated active bone remodelling (as evidenced by the presence of osteoclasts, osteoblasts and calcein uptake). Restricting bone implants to the use of subchondral bone and introduction of cancer cells via intraductal injection maximised metastasis to human bone implants. MDA-MB-231 cells specifically metastasised to human bone (70% metastases) whereas T47D, MCF7, BB3RC32, BB2RC08, and BB6RC37 cells metastasised to both human bone and mouse bones. Importantly, human bone was the preferred metastatic site especially from ER+ PDX (100% metastasis human bone compared with 20–75% to mouse bone), whereas ER-ve PDX developed metastases in 20% of human and 20% of mouse bone. Breast cancer cells underwent a series of molecular changes as they progressed from primary tumours to bone metastasis including altered expression of IL-1B, IL-1R1, S100A4, CTSK, SPP1 and RANK. Inhibiting IL-1B signalling significantly reduced bone metastasis. Conclusions Our reliable and clinically relevant humanised mouse models provide significant advancements in modelling of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Diane Lefley
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Faith Howard
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Fawaz Arshad
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Steven Bradbury
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Hannah Brown
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Claudia Tulotta
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Rachel Eyre
- Manchester Breast Centre, Oglesby Cancer Research Building, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Denis Alférez
- Manchester Breast Centre, Oglesby Cancer Research Building, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - J Mark Wilkinson
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Ingunn Holen
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Robert B Clarke
- Manchester Breast Centre, Oglesby Cancer Research Building, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Penelope Ottewell
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|
12
|
Hurley SK, Cutrone NM, Fath KR, Pajovich HT, Garcia J, Smith AM, Banerjee IA. Self-assembled phenylisoxazole-peptide hybrid assemblies and their interactions with breast and ovarian tumor cells. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2018.1525542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Sara K. Hurley
- Department of Chemistry, Fordham University, Bronx, NY, USA
| | | | - Karl R. Fath
- Department of Biology, Queens College, City University of New York, New York, NY, USA
| | | | - Jeremy Garcia
- Department of Biology, Queens College, City University of New York, New York, NY, USA
| | | | | |
Collapse
|
13
|
Popielarski M, Ponamarczuk H, Stasiak M, Watała C, Świątkowska M. Modifications of disulfide bonds in breast cancer cell migration and invasiveness. Am J Cancer Res 2019; 9:1554-1582. [PMID: 31497343 PMCID: PMC6727000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/22/2019] [Indexed: 06/10/2023] Open
Abstract
Cancer metastasis involves the adhesion of cancer cells to the endothelium. This process can be mediated by integrins which are surface receptors responsible for interactions with ECM proteins. Integrins β1 and αVβ3 represent factors are involved in cancer progression and metastasis. Activation of integrins can be promoted by thiol-disulfide exchanges initiated by Protein Disulfide Isomerase (PDI). The purpose of this study was to prove the involvement of disulfide rearrangements in the molecules of integrins in the course of cancer cell adhesion and migration through the endothelium. We present the evidence which proves that highly metastatic MDA-MB-231 breast cancer cell lines adhere to endothelial cells are more effective than non-invasive MCF-10A and MCF-7 cell lines and that the attachment of MDA-MB-231 to the endothelium can be attenuated either by the agents blocking free thiol groups (DTNB, cystamine or PCMBS) or by PDI inhibitors (Q3Rut, 16F16 or PACMA-31). Furthermore, we prove that the transendothelial migration of MDA-MB-231 cells and contraction of collagen can be blocked by thiol blockers or PDI inhibitors and that these factors affect exposition of free thiols on integrin molecules.
Collapse
Affiliation(s)
- Marcin Popielarski
- Department of Cytobiology and Proteomics, Medical University of Lodz6/8 Mazowiecka St., 92-215 Lodz, Poland
| | - Halszka Ponamarczuk
- Department of Cytobiology and Proteomics, Medical University of Lodz6/8 Mazowiecka St., 92-215 Lodz, Poland
| | - Marta Stasiak
- Department of Cytobiology and Proteomics, Medical University of Lodz6/8 Mazowiecka St., 92-215 Lodz, Poland
| | - Cezary Watała
- Department of Haemostatic Disorders, Medical University of Lodz6/8 Mazowiecka St., 92-215 Lodz, Poland
| | - Maria Świątkowska
- Department of Cytobiology and Proteomics, Medical University of Lodz6/8 Mazowiecka St., 92-215 Lodz, Poland
| |
Collapse
|
14
|
Peláez R, Pariente A, Pérez-Sala Á, Larrayoz IM. Integrins: Moonlighting Proteins in Invadosome Formation. Cancers (Basel) 2019; 11:cancers11050615. [PMID: 31052560 PMCID: PMC6562994 DOI: 10.3390/cancers11050615] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/24/2022] Open
Abstract
Invadopodia are actin-rich protrusions developed by transformed cells in 2D/3D environments that are implicated in extracellular matrix (ECM) remodeling and degradation. These structures have an undoubted association with cancer invasion and metastasis because invadopodium formation in vivo is a key step for intra/extravasation of tumor cells. Invadopodia are closely related to other actin-rich structures known as podosomes, which are typical structures of normal cells necessary for different physiological processes during development and organogenesis. Invadopodia and podosomes are included in the general term 'invadosomes,' as they both appear as actin puncta on plasma membranes next to extracellular matrix metalloproteinases, although organization, regulation, and function are slightly different. Integrins are transmembrane proteins implicated in cell-cell and cell-matrix interactions and other important processes such as molecular signaling, mechano-transduction, and cell functions, e.g., adhesion, migration, or invasion. It is noteworthy that integrin expression is altered in many tumors, and other pathologies such as cardiovascular or immune dysfunctions. Over the last few years, growing evidence has suggested a role of integrins in the formation of invadopodia. However, their implication in invadopodia formation and adhesion to the ECM is still not well known. This review focuses on the role of integrins in invadopodium formation and provides a general overview of the involvement of these proteins in the mechanisms of metastasis, taking into account classic research through to the latest and most advanced work in the field.
Collapse
Affiliation(s)
- Rafael Peláez
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Ana Pariente
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Álvaro Pérez-Sala
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| |
Collapse
|
15
|
Zhan FK, Liu JC, Cheng B, Liu YC, Lai TS, Lin HC, Yeh MY. Tumor targeting with DGEA peptide ligands: a new aromatic peptide amphiphile for imaging cancers. Chem Commun (Camb) 2019; 55:1060-1063. [DOI: 10.1039/c8cc08679f] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A novel AIE-active self-assembled bioprobe TPE-FDGEA has been developed for selective cancer cell imaging.
Collapse
Affiliation(s)
- Fu-Kai Zhan
- Department of Materials Science and Engineering
- National Chiao Tung University
- Hsinchu
- Taiwan
| | - Jyun-Cheng Liu
- Department of Chemistry
- Chung Yuan Christian University
- Taoyuan
- Taiwan
| | - Bill Cheng
- Department of Materials Science and Engineering
- National Chiao Tung University
- Hsinchu
- Taiwan
| | - Yen-Chu Liu
- Department of Materials Science and Engineering
- National Chiao Tung University
- Hsinchu
- Taiwan
| | - Tsung-Sheng Lai
- Department of Materials Science and Engineering
- National Chiao Tung University
- Hsinchu
- Taiwan
| | - Hsin-Chieh Lin
- Department of Materials Science and Engineering
- National Chiao Tung University
- Hsinchu
- Taiwan
| | - Mei-Yu Yeh
- Department of Chemistry
- Chung Yuan Christian University
- Taoyuan
- Taiwan
| |
Collapse
|
16
|
Adorno-Cruz V, Liu H. Regulation and functions of integrin α2 in cell adhesion and disease. Genes Dis 2018; 6:16-24. [PMID: 30906828 PMCID: PMC6411621 DOI: 10.1016/j.gendis.2018.12.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/24/2018] [Indexed: 12/23/2022] Open
Abstract
Integrins are cell adhesion molecules that are composed of an alpha (α) subunit and a beta (β) subunit with affinity for different extracellular membrane components. The integrin family includes 24 known members that actively regulate cellular growth, differentiation, and apoptosis. Each integrin heterodimer has a particular function in defined contexts as well as some partially overlapping features with other members in the family. As many reviews have covered the general integrin family in molecular and cellular studies in life science, this review will focus on the specific regulation, function, and signaling of integrin α2 subunit (CD49b, VLA-2; encoded by the gene ITGA2) in partnership with β1 (CD29) subunit in normal and cancer cells. Its roles in cell adhesion, cell motility, angiogenesis, stemness, and immune/blood cell regulations are discussed. The pivotal role of integrin α2 in many diseases such as cancer suggests its potential to be used as a novel therapeutic target.
Collapse
Affiliation(s)
- Valery Adorno-Cruz
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Pharmacology Graduate Program, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Huiping Liu
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Medicine, Hematology/Oncology Division, Northwestern University, Chicago, IL 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
17
|
The vacuolar-type ATPase inhibitor archazolid increases tumor cell adhesion to endothelial cells by accumulating extracellular collagen. PLoS One 2018; 13:e0203053. [PMID: 30204757 PMCID: PMC6133348 DOI: 10.1371/journal.pone.0203053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/14/2018] [Indexed: 11/19/2022] Open
Abstract
The vacuolar-type H+-ATPase (v-ATPase) is the major proton pump that acidifies intracellular compartments of eukaryotic cells. Since the inhibition of v-ATPase resulted in anti-tumor and anti-metastatic effects in different tumor models, this enzyme has emerged as promising strategy against cancer. Here, we used the well-established v-ATPase inhibitor archazolid, a natural product first isolated from the myxobacterium Archangium gephyra, to study the consequences of v-ATPase inhibition in endothelial cells (ECs), in particular on the interaction between ECs and cancer cells, which has been neglected so far. Human endothelial cells treated with archazolid showed an increased adhesion of tumor cells, whereas the transendothelial migration of tumor cells was reduced. The adhesion process was independent from the EC adhesion molecules ICAM-1, VCAM-1, E-selectin and N-cadherin. Instead, the adhesion was mediated by β1-integrins expressed on tumor cells, as blocking of the integrin β1 subunit reversed this process. Tumor cells preferentially adhered to the β1-integrin ligand collagen and archazolid led to an increase in the amount of collagen on the surface of ECs. The accumulation of collagen was accompanied by a strong decrease of the expression and activity of the protease cathepsin B. Overexpression of cathepsin B in ECs prevented the capability of archazolid to increase the adhesion of tumor cells onto ECs. Our study demonstrates that the inhibition of v-ATPase by archazolid induces a pro-adhesive phenotype in endothelial cells that promotes their interaction with cancer cells, whereas the transmigration of tumor cells was reduced. These findings further support archazolid as a promising anti-metastatic compound.
Collapse
|
18
|
Casal JI, Bartolomé RA. RGD cadherins and α2β1 integrin in cancer metastasis: A dangerous liaison. Biochim Biophys Acta Rev Cancer 2018; 1869:321-332. [PMID: 29673969 DOI: 10.1016/j.bbcan.2018.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 12/24/2022]
Abstract
We propose a new cadherin family classification comprising epithelial cadherins (cadherin 17 [CDH17], cadherin 16, VE-cadherin, cadherin 6 and cadherin 20) containing RGD motifs within their sequences. Expression of some RGD cadherins is associated with aggressive forms of cancer during the late stages of metastasis, and CDH17 and VE-cadherin have emerged as critical actors in cancer metastasis. After binding to α2β1 integrin, these cadherins promote integrin β1 activation, and thereby cell adhesion, invasion and proliferation, in liver and lung metastasis. Activation of α2β1 integrin provokes an affinity increase for type IV collagen, a major component of the basement membrane and a critical partner for cell anchoring in liver and other metastatic organs. Activation of α2β1 integrin by RGD motifs breaks an old paradigm of integrin classification and supports an important role of this integrin in cancer metastasis. Recently, synthetic peptides containing the RGD motif of CDH17 elicited highly specific and selective antibodies that block the ability of CDH17 RGD to activate α2β1 integrin. These monoclonal antibodies inhibit metastatic colonization in orthotopic mouse models of liver and lung metastasis for colorectal cancer and melanoma, respectively. Hopefully, blocking the cadherin RGD ligand capacity will give us control over the integrin activity in solid tumors metastasis, paving the way for development of new agents of cancer treatment.
Collapse
Affiliation(s)
- J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039 Madrid, Spain.
| | - Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039 Madrid, Spain
| |
Collapse
|
19
|
Bartolomé RA, Torres S, Isern de Val S, Escudero-Paniagua B, Calviño E, Teixidó J, Casal JI. VE-cadherin RGD motifs promote metastasis and constitute a potential therapeutic target in melanoma and breast cancers. Oncotarget 2018; 8:215-227. [PMID: 27966446 PMCID: PMC5352113 DOI: 10.18632/oncotarget.13832] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/14/2016] [Indexed: 12/15/2022] Open
Abstract
We have investigated the role of vascular-endothelial (VE)-cadherin in melanoma and breast cancer metastasis. We found that VE-cadherin is expressed in highly aggressive melanoma and breast cancer cell lines. Remarkably, inactivation of VE-cadherin triggered a significant loss of malignant traits (proliferation, adhesion, invasion and transendothelial migration) in melanoma and breast cancer cells. These effects, except transendothelial migration, were induced by the VE-cadherin RGD motifs. Co-immunoprecipitation experiments demonstrated an interaction between VE-cadherin and α2β1 integrin, with the RGD motifs found to directly affect β1 integrin activation. VE-cadherin-mediated integrin signaling occurred through specific activation of SRC, ERK and JNK, including AKT in melanoma. Knocking down VE-cadherin suppressed lung colonization capacity of melanoma or breast cancer cells inoculated in mice, while pre-incubation with VE-cadherin RGD peptides promoted lung metastasis for both cancer types. Finally, an in silico study revealed the association of high VE-cadherin expression with poor survival in a subset of melanoma patients and breast cancer patients showing low CD34 expression. These findings support a general role for VE-cadherin and other RGD cadherins as critical regulators of lung and liver metastasis in multiple solid tumours. These results pave the way for cadherin-specific RGD targeted therapies to control disseminated metastasis in multiple cancers.
Collapse
Affiliation(s)
- Rubén A Bartolomé
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Ramiro de Maeztu, Madrid, Spain
| | - Sofía Torres
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Ramiro de Maeztu, Madrid, Spain
| | - Soledad Isern de Val
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Ramiro de Maeztu, Madrid, Spain
| | - Beatriz Escudero-Paniagua
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Ramiro de Maeztu, Madrid, Spain
| | - Eva Calviño
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Ramiro de Maeztu, Madrid, Spain
| | - Joaquín Teixidó
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Ramiro de Maeztu, Madrid, Spain
| | - J Ignacio Casal
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Ramiro de Maeztu, Madrid, Spain
| |
Collapse
|
20
|
Collateral Damage Intended-Cancer-Associated Fibroblasts and Vasculature Are Potential Targets in Cancer Therapy. Int J Mol Sci 2017; 18:ijms18112355. [PMID: 29112161 PMCID: PMC5713324 DOI: 10.3390/ijms18112355] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/25/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023] Open
Abstract
After oncogenic transformation, tumor cells rewire their metabolism to obtain sufficient energy and biochemical building blocks for cell proliferation, even under hypoxic conditions. Glucose and glutamine become their major limiting nutritional demands. Instead of being autonomous, tumor cells change their immediate environment not only by their metabolites but also by mediators, such as juxtacrine cell contacts, chemokines and other cytokines. Thus, the tumor cells shape their microenvironment as well as induce resident cells, such as fibroblasts and endothelial cells (ECs), to support them. Fibroblasts differentiate into cancer-associated fibroblasts (CAFs), which produce a qualitatively and quantitatively different extracellular matrix (ECM). By their contractile power, they exert tensile forces onto this ECM, leading to increased intratumoral pressure. Moreover, along with enhanced cross-linkage of the ECM components, CAFs thus stiffen the ECM. Attracted by tumor cell- and CAF-secreted vascular endothelial growth factor (VEGF), ECs sprout from pre-existing blood vessels during tumor-induced angiogenesis. Tumor vessels are distinct from EC-lined vessels, because tumor cells integrate into the endothelium or even mimic and replace it in vasculogenic mimicry (VM) vessels. Not only the VM vessels but also the characteristically malformed EC-lined tumor vessels are typical for tumor tissue and may represent promising targets in cancer therapy.
Collapse
|
21
|
Crowley VM, Huard DJE, Lieberman RL, Blagg BSJ. Second Generation Grp94-Selective Inhibitors Provide Opportunities for the Inhibition of Metastatic Cancer. Chemistry 2017; 23:15775-15782. [PMID: 28857290 DOI: 10.1002/chem.201703398] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Indexed: 12/24/2022]
Abstract
Glucose regulated protein 94 (Grp94) is the endoplasmic reticulum (ER) resident isoform of the 90 kDa heat shock protein (Hsp90) family and its inhibition represents a promising therapeutic target for the treatment of many diseases. Modification of the first generation cis-amide bioisostere imidazole to alter the angle between the resorcinol ring and the benzyl side chain via cis-amide replacements produced compounds with improved Grp94 affinity and selectivity. Structure-activity relationship studies led to the discovery of compound 30, which exhibits 540 nm affinity and 73-fold selectivity towards Grp94. Grp94 is responsible for the maturation and trafficking of proteins associated with cell signaling and motility, including select integrins. The Grp94-selective inhibitor 30 was shown to exhibit potent anti-migratory effects against multiple aggressive and metastatic cancers.
Collapse
Affiliation(s)
- Vincent M Crowley
- Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Dr. Malott 4070, Lawrence, KS, 66045, USA
| | - Dustin J E Huard
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Raquel L Lieberman
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Brian S J Blagg
- Warren Family Research Center for Drug Discovery and Development, and Department of Chemistry & Biochemistry, University of Notre Dame, 305 McCourtney Hall, Notre Dame, IN, 46556, USA
| |
Collapse
|
22
|
Mashreghi M, Azarpara H, Bazaz MR, Jafari A, Masoudifar A, Mirzaei H, Jaafari MR. Angiogenesis biomarkers and their targeting ligands as potential targets for tumor angiogenesis. J Cell Physiol 2017; 233:2949-2965. [DOI: 10.1002/jcp.26049] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/12/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Mohammad Mashreghi
- NanotechnologyResearch Center; Mashhad University of Medical Sciences; Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Mashhad Iran
| | - Hassan Azarpara
- School of Medicine; Iran University of Medical Sciences; Tehran Iran
| | - Mahere R. Bazaz
- Division of Biotechnology, Faculty of Veterinary Medicine; Ferdowsi University of Mashhad; Mashhad Iran
| | - Arash Jafari
- School of Medicine; Birjand University of Medical Sciences; Birjand Iran
| | - Aria Masoudifar
- Department of Molecular Biotechnology, Cell Science Research Center; Royan Institute for Biotechnology; ACECR Isfahan Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mahmoud R. Jaafari
- NanotechnologyResearch Center; Mashhad University of Medical Sciences; Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
23
|
A Novel Three-Dimensional Platform to Investigate Neoangiogenesis, Transendothelial Migration, and Metastasis of MDAMB-231 Breast Cancer Cells. Plast Reconstr Surg 2017; 138:472e-482e. [PMID: 27556622 DOI: 10.1097/prs.0000000000002470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND A crucial step in the progression of cancer involves the transendothelial migration of tumor cells into the bloodstream and invasion at distant sites. Most in vitro models of malignant cell behavior do not account for the presence of and interaction with vascular cells. Three-dimensional platforms to further explore the factors responsible for metastatic cellular behavior are under intensive investigation. METHODS Hydrogels with encapsulated MDAMB-231 breast cancer cells were fabricated with a central microchannel. The microchannel was lined with a co-culture of human umbilical vein endothelial cells and human aortic smooth muscle cells. For comparison, co-culture-seeded microchannels without breast cancer cells (MDAMB-negative) were fabricated. RESULTS After 7 and 14 days, the endoluminal lining of encapsulated MDAMB-231 co-culture-seeded microchannels demonstrated aberrant endothelial cell and smooth muscle cell organization and breast cancer cell transendothelial migration. MDAMB-231 cells performed matrix remodeling, forming tumor aggregates within the bulk, migrating preferentially toward the hydrogel "neovessel." In contrast, MDAMB-negative constructs demonstrated maintenance of an intact endoluminal lining composed of endothelial cells and smooth muscle cells that organized into discrete layers. Furthermore, the thicknesses of the endoluminal lining of MDAMB-negative constructs were significantly greater than encapsulated MDAMB-231 co-culture-seeded constructs after 7 and 14 days (p = 0.012 and p < 0.001, respectively). CONCLUSION The authors have created a powerful tool that may have tremendous impact on furthering our understanding of cancer recurrence and metastasis, shedding light on these poorly understood phenomena.
Collapse
|
24
|
Aragon-Sanabria V, Pohler SE, Eswar VJ, Bierowski M, Gomez EW, Dong C. VE-Cadherin Disassembly and Cell Contractility in the Endothelium are Necessary for Barrier Disruption Induced by Tumor Cells. Sci Rep 2017; 7:45835. [PMID: 28393886 PMCID: PMC5385522 DOI: 10.1038/srep45835] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/03/2017] [Indexed: 12/20/2022] Open
Abstract
During metastasis, breakdown of the endothelial barrier is critical for tumor cell extravasation through blood vessel walls and is mediated by a combination of tumor secreted soluble factors and receptor-ligand interactions. However, a complete mechanism governing tumor cell transendothelial migration remains unclear. Here, we investigate the roles of tumor-associated signals in regulating endothelial cell contractility and adherens junction disassembly leading to endothelial barrier breakdown. We show that Src mediates VE-cadherin disassembly in response to metastatic melanoma cells. Through the use of pharmacological inhibitors of cytoskeletal contractility we find that endothelial cell contractility is responsive to interactions with metastatic cancer cells and that reducing endothelial cell contractility abrogates migration of melanoma cells across endothelial monolayers. Furthermore, we find that a combination of tumor secreted soluble factors and receptor-ligand interactions mediate activation of Src within endothelial cells that is necessary for phosphorylation of VE-cadherin and for breakdown of the endothelial barrier. Together, these results provide insight into how tumor cell signals act in concert to modulate cytoskeletal contractility and adherens junctions disassembly during extravasation and may aid in identification of therapeutic targets to block metastasis.
Collapse
Affiliation(s)
- Virginia Aragon-Sanabria
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| | - Steven E. Pohler
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Vikram J. Eswar
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| | - Matthew Bierowski
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Esther W. Gomez
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Cheng Dong
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| |
Collapse
|
25
|
Breast cancer-secreted miR-939 downregulates VE-cadherin and destroys the barrier function of endothelial monolayers. Cancer Lett 2016; 384:94-100. [PMID: 27693459 DOI: 10.1016/j.canlet.2016.09.013] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 11/21/2022]
Abstract
Exosomes-secreted microRNAs play an important role in metastatic spread. During this process breast cancer cells acquire the ability to transmigrate through blood vessels by inducing changes in the endothelial barrier. We focused on miR-939 that is predicted to target VE-cadherin, a component of adherens junction involved in vessel permeability. By in silico analysis miR-939 was found highly expressed in the basal-like tumor subtypes and in our cohort of 63 triple-negative breast cancers (TNBCs) its expression significantly interacted with lymph node status in predicting disease-free survival probability. We demonstrated, in vitro, that miR-939 directly targets VE-cadherin leading to an increase in HUVECs monolayer permeability. MDA-MB-231 cells transfected with a miR-939 mimic, released miR-939 in exosomes that, once internalized in endothelial cells, favored trans-endothelial migration of MDA-MB-231-GFP cells by the disruption of the endothelial barrier. Notably, when up taken in endothelial cells exosomes caused VE-cadherin down-regulation specifically through miR-939 as we demonstrated by inhibiting miR-939 expression in exosomes-releasing TNBC cells. Together, our data indentify an extracellular pro-tumorigenic role for tumor-derived, exosome-associated miR-939 that can explain its association with worse prognosis in TNBCs.
Collapse
|
26
|
Häuselmann I, Roblek M, Protsyuk D, Huck V, Knopfova L, Grässle S, Bauer AT, Schneider SW, Borsig L. Monocyte Induction of E-Selectin-Mediated Endothelial Activation Releases VE-Cadherin Junctions to Promote Tumor Cell Extravasation in the Metastasis Cascade. Cancer Res 2016; 76:5302-12. [PMID: 27488527 DOI: 10.1158/0008-5472.can-16-0784] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/27/2016] [Indexed: 12/24/2022]
Abstract
Tumor cells interact with blood constituents and these interactions promote metastasis. Selectins are vascular receptors facilitating interactions of tumor cells with platelets, leukocytes, and endothelium, but the role of endothelial E-selectin remains unclear. Here we show that E-selectin is a major receptor for monocyte recruitment to tumor cell-activated endothelium. Experimental and spontaneous lung metastasis using murine tumor cells, without E-selectin ligands, were attenuated in E-selectin-deficient mice. Tumor cell-derived CCL2 promoted endothelial activation, resulting in enhanced endothelial E-selectin expression. The recruitment of inflammatory monocytes to metastasizing tumor cells was dependent on the local endothelial activation and the presence of E-selectin. Monocytes promoted transendothelial migration of tumor cells through the induction of E-selectin-dependent endothelial retractions and a subsequent modulation of tight junctions through dephosphorylation of VE-cadherin. Thus, endothelial E-selectin shapes the tumor microenvironment through the recruitment, adhesion, and activation of monocytes that facilitate tumor cell extravasation and thereby metastasis. These findings provide evidence that endothelial E-selectin is a novel factor contributing to endothelial retraction required for efficient lung metastasis. Cancer Res; 76(18); 5302-12. ©2016 AACR.
Collapse
Affiliation(s)
- Irina Häuselmann
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Marko Roblek
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Darya Protsyuk
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Volker Huck
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lucia Knopfova
- International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital and Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Sandra Grässle
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander T Bauer
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan W Schneider
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lubor Borsig
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zurich, Switzerland.
| |
Collapse
|
27
|
Higuchi K, Inokuchi M, Takagi Y, Ishikawa T, Otsuki S, Uetake H, Kojima K, Kawano T. Cadherin 5 expression correlates with poor survival in human gastric cancer. J Clin Pathol 2016; 70:217-221. [PMID: 27466381 DOI: 10.1136/jclinpath-2016-203640] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 06/29/2016] [Accepted: 07/02/2016] [Indexed: 02/06/2023]
Abstract
AIMS Although expressed in tumour cells of various malignancies, cadherin 5 (CDH5), also known as vascular endothelial cadherin, plays an important role in homotypic cell-cell adhesion among epithelial cells. However, the clinical significance of CDH5 expression in gastric cancer has not been sufficiently demonstrated. In this study, CDH5 expression in gastric cancer was evaluated and the correlations between CDH5 expression and the clinicopathological features and outcomes of the disease were examined. METHODS Differentiated-type gastric adenocarcinomas obtained from 102 patients who underwent gastrectomy were analysed. CDH5 expression was assessed by immunohistochemical staining of the membranes of the cancer cells. RESULTS High CDH5 expression was significantly associated with the following clinicopathological variables related to tumour progression: depth of invasion (p=0.012), venous invasion (p=0.013), lymphatic invasion (p=0.001), metastatic lymph nodes (p=0.009), pathological stage (p=0.008) and distant metastasis or recurrent disease (p=0.009). Patients with high CDH5 expression had significantly poorer disease-specific survival (p=0.021), although CDH5 was not determined to be an independent prognostic factor by multivariate analysis. CONCLUSIONS CDH5 may play a key role in the progression or metastasis of differentiated-type gastric cancer and serve as a target for its treatment.
Collapse
Affiliation(s)
- Kyoko Higuchi
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mikito Inokuchi
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoko Takagi
- Department of Surgical Specialties, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshiaki Ishikawa
- Department of Surgical Specialties, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sho Otsuki
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Uetake
- Department of Surgical Specialties, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuyuki Kojima
- Department of Minimally Invasive Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatsuyuki Kawano
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
28
|
Crowley VM, Khandelwal A, Mishra S, Stothert AR, Huard DJE, Zhao J, Muth A, Duerfeldt AS, Kizziah JL, Lieberman RL, Dickey CA, Blagg BSJ. Development of Glucose Regulated Protein 94-Selective Inhibitors Based on the BnIm and Radamide Scaffold. J Med Chem 2016; 59:3471-88. [PMID: 27003516 DOI: 10.1021/acs.jmedchem.6b00085] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glucose regulated protein 94 (Grp94) is the endoplasmic reticulum resident of the heat shock protein 90 kDa (Hsp90) family of molecular chaperones. Grp94 associates with many proteins involved in cell adhesion and signaling, including integrins, Toll-like receptors, immunoglobulins, and mutant myocilin. Grp94 has been implicated as a target for several therapeutic areas including glaucoma, cancer metastasis, and multiple myeloma. While 85% identical to other Hsp90 isoforms, the N-terminal ATP-binding site of Grp94 possesses a unique hydrophobic pocket that was used to design isoform-selective inhibitors. Incorporation of a cis-amide bioisostere into the radamide scaffold led to development of the original Grp94-selective inhibitor, BnIm. Structure-activity relationship studies have now been performed on the aryl side chain of BnIm, which resulted in improved analogues that exhibit better potency and selectivity for Grp94. These analogues also manifest superior antimigratory activity in a metastasis model as well as enhanced mutant myocilin degradation in a glaucoma model compared to BnIm.
Collapse
Affiliation(s)
- Vincent M Crowley
- Department of Medicinal Chemistry, The University of Kansas , 1251 Wescoe Hall Drive, Malott Hall 4070, Lawrence, Kansas 66045-7563, United States
| | - Anuj Khandelwal
- Department of Medicinal Chemistry, The University of Kansas , 1251 Wescoe Hall Drive, Malott Hall 4070, Lawrence, Kansas 66045-7563, United States
| | - Sanket Mishra
- Department of Medicinal Chemistry, The University of Kansas , 1251 Wescoe Hall Drive, Malott Hall 4070, Lawrence, Kansas 66045-7563, United States
| | - Andrew R Stothert
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, University of South Florida , Tampa, Florida 33613, United States
| | - Dustin J E Huard
- School of Chemistry and Biochemistry, Georgia Institute of Technology , Atlanta, Georgia 30332 United States
| | - Jinbo Zhao
- Department of Medicinal Chemistry, The University of Kansas , 1251 Wescoe Hall Drive, Malott Hall 4070, Lawrence, Kansas 66045-7563, United States
| | - Aaron Muth
- Department of Medicinal Chemistry, The University of Kansas , 1251 Wescoe Hall Drive, Malott Hall 4070, Lawrence, Kansas 66045-7563, United States
| | - Adam S Duerfeldt
- Department of Medicinal Chemistry, The University of Kansas , 1251 Wescoe Hall Drive, Malott Hall 4070, Lawrence, Kansas 66045-7563, United States
| | - James L Kizziah
- School of Chemistry and Biochemistry, Georgia Institute of Technology , Atlanta, Georgia 30332 United States
| | - Raquel L Lieberman
- School of Chemistry and Biochemistry, Georgia Institute of Technology , Atlanta, Georgia 30332 United States
| | - Chad A Dickey
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, University of South Florida , Tampa, Florida 33613, United States
| | - Brian S J Blagg
- Department of Medicinal Chemistry, The University of Kansas , 1251 Wescoe Hall Drive, Malott Hall 4070, Lawrence, Kansas 66045-7563, United States
| |
Collapse
|
29
|
Chen MB, Lamar JM, Li R, Hynes RO, Kamm RD. Elucidation of the Roles of Tumor Integrin β1 in the Extravasation Stage of the Metastasis Cascade. Cancer Res 2016; 76:2513-24. [PMID: 26988988 DOI: 10.1158/0008-5472.can-15-1325] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 02/27/2016] [Indexed: 12/22/2022]
Abstract
Tumor integrin β1 (ITGB1) contributes to primary tumor growth and metastasis, but its specific roles in extravasation have not yet been clearly elucidated. In this study, we engineered a three-dimensional microfluidic model of the human microvasculature to recapitulate the environment wherein extravasation takes place and assess the consequences of β1 depletion in cancer cells. Combined with confocal imaging, these tools allowed us to decipher the detailed morphology of transmigrating tumor cells and associated endothelial cells in vitro at high spatio-temporal resolution not easily achieved in conventional transmigration assays. Dynamic imaging revealed that β1-depleted cells lacked the ability to sustain protrusions into the subendothelial matrix in contrast with control cells. Specifically, adhesion via α3β1 and α6β1 to subendothelial laminin was a critical prerequisite for successful transmigration. β1 was required to invade past the endothelial basement membrane, whereas its attenuation in a syngeneic tumor model resulted in reduced metastatic colonization of the lung, an effect not observed upon depletion of other integrin alpha and beta subunits. Collectively, our findings in this novel model of the extravasation microenvironment revealed a critical requirement for β1 in several steps of extravasation, providing new insights into the mechanisms underlying metastasis. Cancer Res; 76(9); 2513-24. ©2016 AACR.
Collapse
Affiliation(s)
- Michelle B Chen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - John M Lamar
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ran Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Richard O Hynes
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts. Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
30
|
Whom to blame for metastasis, the epithelial-mesenchymal transition or the tumor microenvironment? Cancer Lett 2016; 380:359-68. [PMID: 26791236 DOI: 10.1016/j.canlet.2015.12.033] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/22/2015] [Accepted: 12/25/2015] [Indexed: 02/06/2023]
Abstract
Changes in the tumor microenvironment (TME) can trigger the activation of otherwise non-malignant cells to become highly aggressive and motile. This is evident during initial tumor growth when the poor vascularization in tumors generates hypoxic regions that trigger the latent embryonic program, epithelial-to-mesenchymal transition (EMT), in epithelial carcinoma cells (e-cars) leading to highly motile mesenchymal-like carcinoma cells (m-cars), which also acquire cancer stem cell properties. After that, specific bidirectional interactions take place between m-cars and the cellular components of TME at different stages of metastasis. These interactions include several vicious positive feedback loops in which m-cars trigger a phenotypic switch, causing normal stromal cells to become pro-tumorigenic, which then further promote the survival, motility, and proliferation of m-cars. Accordingly, there is not a single culprit accounting for metastasis. Instead both m-cars and the TME dynamically interact, evolve and promote metastasis. In this review, we discuss the current status of the known interactions between m-cars and the TME during different stages of metastasis and how these interactions promote the metastatic activity of highly malignant m-cars by promoting their invasive mesenchymal phenotype and CSC properties.
Collapse
|
31
|
Naci D, Vuori K, Aoudjit F. Alpha2beta1 integrin in cancer development and chemoresistance. Semin Cancer Biol 2015; 35:145-53. [PMID: 26297892 DOI: 10.1016/j.semcancer.2015.08.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/10/2015] [Accepted: 08/14/2015] [Indexed: 01/06/2023]
Abstract
Extracellular matrix, via its receptors the integrins, has emerged as a crucial factor in cancer development. The α2β1 integrin is a major collagen receptor that is widely expressed and known to promote cell migration and control tissue homeostasis. Growing evidence suggests that it can be a key pathway in cancer. Recent studies have shown that α2β1 integrin is a regulator of cancer metastasis either by promoting or inhibiting the dissemination process of cancer cells. The α2β1 integrin signaling can also enhance tumor angiogenesis. Emerging evidence supports a role for α2β1 integrin in cancer chemoresistance especially in hematological malignancies originating from the T cell lineage. In addition, α2β1 integrin has been associated with cancer stem cells. In this review, we will discuss the complex role of α2β1 integrin in these processes. Collagen is a major matrix protein of the tumor microenvironment and thus, understanding how α2β1 integrin regulates cancer pathogenesis is likely to lead to new therapeutic approaches and agents for cancer treatment.
Collapse
Affiliation(s)
- Dalila Naci
- Centre de recherche du CHU de Québec, Axe des maladies infectieuses et immunitaires and Département de Microbiologie-Immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| | - Kristiina Vuori
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Fawzi Aoudjit
- Centre de recherche du CHU de Québec, Axe des maladies infectieuses et immunitaires and Département de Microbiologie-Immunologie, Faculté de Médecine, Université Laval, Québec, Canada.
| |
Collapse
|
32
|
Mittal M, Urao N, Hecquet CM, Zhang M, Sudhahar V, Gao XP, Komarova Y, Ushio-Fukai M, Malik AB. Novel role of reactive oxygen species-activated Trp melastatin channel-2 in mediating angiogenesis and postischemic neovascularization. Arterioscler Thromb Vasc Biol 2015; 35:877-87. [PMID: 25675998 DOI: 10.1161/atvbaha.114.304802] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Transient receptor potential melastatin-2 (TRPM2) channel is a nonselective cation channel that mediates influx of Ca(2+) and Na(+) with relative permeability of PCa:PNa ≈0.6 in response to cellular oxidative stress. As angiogenesis and ischemic neovascularization are both significantly dependent on oxidant signaling, here we investigated the possible role of vascular endothelial growth factor (VEGF)-induced reactive oxygen species production in activating TRPM2-dependent Ca(2+) signaling and in the mechanism of angiogenesis and ischemic neovascularization. APPROACH AND RESULTS We observed that VEGF stimulation rapidly induced the association of TRPM2 and cellular Src kinase with vascular endothelial-cadherin forming a signalplex at vascular endothelial-cadherin junctions in endothelial cells. Using endothelial cells isolated from TRPM2(-/-) mice or after small interfering RNA depletion of TRPM2, we demonstrated that TRPM2-activated Ca(2+) signaling was required for cellular Src kinase-induced phosphorylation of vascular endothelial-cadherin at Y658 and Y731, the crucial sites involved in vascular endothelial-cadherin internalization in response to VEGF. VEGF-induced reactive oxygen species generation activated TRPM2-induced Ca(2+) entry, whereas the reactive oxygen species-insensitive TRPM2 mutant (C1008→A) showed impaired Ca(2+) entry. Endothelial cells depleted of TRPM2 also displayed significantly perturbed migratory phenotype and impaired activation of cellular Src in response to VEGF. TRPM2(-/-) mice reconstituted with wild-type myeloid cells demonstrated aberrant angiogenesis and neovascularization in the hindlimb ischemia model as compared with wild-type mice. CONCLUSIONS VEGF-induced angiogenesis and postischemic neovascularization in mice required reactive oxygen species generation in endothelial cells and resultant TRPM2 activation. Thus, our findings provide novel insight into the role of TRPM2 in mechanism of angiogenesis and ischemic neovascularization.
Collapse
Affiliation(s)
- Manish Mittal
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Norifumi Urao
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Claudie M Hecquet
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Min Zhang
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Varadarajan Sudhahar
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Xiao-Pei Gao
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Yulia Komarova
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Masuko Ushio-Fukai
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Asrar B Malik
- From the Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago.
| |
Collapse
|
33
|
Zeng Z, Inoue K, Sun H, Leng T, Feng X, Zhu L, Xiong ZG. TRPM7 regulates vascular endothelial cell adhesion and tube formation. Am J Physiol Cell Physiol 2014; 308:C308-18. [PMID: 25472964 DOI: 10.1152/ajpcell.00275.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Transient receptor potential melastatin 7 (TRPM7) is a nonselective cation channel with an α-kinase domain in its COOH terminal, known to play a role in diverse physiological and pathological processes such as Mg2+ homeostasis, cell proliferation, and hypoxic neuronal injury. Increasing evidence suggests that TRPM7 contributes to the physiology/pathology of vascular systems. For example, we recently demonstrated that silencing TRPM7 promotes growth and proliferation and protects against hyperglycemia-induced injury in human umbilical vein endothelial cells (HUVECs). Here we investigated the potential effects of TRPM7 on morphology, adhesion, migration, and tube formation of vascular endothelial cells and the potential underlying mechanism. We showed that inhibition of TRPM7 function in HUVECs by silencing TRPM7 decreases the density of TRPM7-like current and cell surface area and inhibits cell adhesion to Matrigel. Silencing TRPM7 also promotes cell migration, wound healing, and tube formation. Further studies showed that the extracellular signal-regulated kinase (ERK) pathway is involved in the change of cell morphology and the increase in HUVEC migration induced by TRPM7 silencing. We also demonstrated that silencing TRPM7 enhances the phosphorylation of myosin light chain (MLC) in HUVECs, which might be involved in the enhancement of cell contractility and motility. Collectively, our data suggest that the TRPM7 channel negatively regulates the function of vascular endothelial cells. Further studies on the underlying mechanism may facilitate the development of the TRPM7 channel as a target for the therapeutic intervention of vascular diseases.
Collapse
Affiliation(s)
- Zhao Zeng
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Ministry of Health Key Laboratory of Thrombosis and Hemostasis, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China; and Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Koichi Inoue
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Huawei Sun
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Tiandong Leng
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Xuechao Feng
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Li Zhu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Ministry of Health Key Laboratory of Thrombosis and Hemostasis, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China; and
| | - Zhi-Gang Xiong
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
34
|
Weidert E, Pohler SE, Gomez EW, Dong C. Actinomyosin contraction, phosphorylation of VE-cadherin, and actin remodeling enable melanoma-induced endothelial cell-cell junction disassembly. PLoS One 2014; 9:e108092. [PMID: 25225982 PMCID: PMC4167543 DOI: 10.1371/journal.pone.0108092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/25/2014] [Indexed: 02/07/2023] Open
Abstract
During melanoma cell extravasation through the vascular endothelium, melanoma cells interact with endothelial cells through secretion of cytokines and by adhesion between proteins displayed on opposing cell surfaces. How these tumor cell associated signals together regulate the dynamics of intracellular signaling pathways within endothelial cells leading to endothelial cell-cell junction disruption is not well understood. Here, we used a combination of experimental and computational approaches to examine the individual and combined effects of activation of the vascular cell adhesion molecule (VCAM)-1, interleukin (IL)-8, and IL-1β signaling pathways on the integrity of vascular junctions. Our simulations predict a multifaceted interplay of signaling resulting from individual activation of VCAM-1, IL-8 and IL-1β pathways that is neither synergistic nor additive compared to all inputs turned on simultaneously. Furthermore, we show that the levels of phosphorylated proteins associated with actinomyosin contractility and junction disassembly peak prior to those related to actin remodeling. The results of this work provide insight into the dynamics of tumor-mediated endothelial junction disassembly and suggest that targeting proteins downstream of several interaction pathways may be the most effective therapeutic approach to reduce melanoma extravasation.
Collapse
Affiliation(s)
- Eric Weidert
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Steven E. Pohler
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Esther W. Gomez
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (EWG); (CD)
| | - Cheng Dong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (EWG); (CD)
| |
Collapse
|
35
|
Haidari M, Zhang W, Willerson JT, Dixon RA. Disruption of endothelial adherens junctions by high glucose is mediated by protein kinase C-β-dependent vascular endothelial cadherin tyrosine phosphorylation. Cardiovasc Diabetol 2014; 13:105. [PMID: 25927959 PMCID: PMC4223716 DOI: 10.1186/1475-2840-13-105] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/29/2014] [Indexed: 12/17/2022] Open
Abstract
Background Hyperglycemia has been recognized as a primary factor in endothelial barrier dysfunction and in the development of micro- and macrovascular diseases associated with diabetes, but the underlying biochemical mechanisms remain elusive. Tyrosine phosphorylation of vascular endothelial cadherin (VE-cad) leads to the disruption of endothelial adherens junctions and increases the transendothelial migration (TEM) of leukocytes. Methods VE-cad tyrosine phosphorylation, adherens junction integrity and TEM of monocytes in human umbilical vein endothelial cells (HUVECs) treated with high-concentration glucose were evaluated. The role of protein kinase C (PKC) in induction of endothelial cells adherence junction disruption by exposure of HUVECs to high concentration of glucose was explored. Results The treatment of HUVEC with high-concentration glucose increased VE-cad tyrosine phosphorylation, whereas mannitol or 3-O-methyl-D-glucose had no effect. In addition, high-concentration glucose increased the dissociation of the VE-cad–β-catenin complex, activation of the Wnt/β-catenin pathway, and the TEM of monocytes. These alterations were accompanied by the activation of endothelial PKC and increased phosphorylation of ERK and myosin light chain (MLC). High-concentration glucose-induced tyrosine phosphorylation of VE-cad was attenuated by: 1- the inhibition of PKC-β by overexpression of dominant-negative PKC-β 2- inhibition of MLC phosphorylation by overexpression of a nonphosphorylatable dominant-negative form of MLC, 3- the inhibition of actin polymerization by cytochalasin D and 4- the treatment of HUVECs with forskolin (an activator of adenylate cyclase). Conclusions Our findings show that the high-concentration glucose-induced disruption of endothelial adherens junctions is mediated by tyrosine phosphorylation of VE-cad through PKC-β and MLC phosphorylation.
Collapse
Affiliation(s)
- Mehran Haidari
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 77030, Houston, TX, USA. .,Texas Heart Institute at St. Luke's Episcopal Hospital, PO Box 20345 C1000, 77030, Houston, TX, USA.
| | - Wei Zhang
- Texas Heart Institute at St. Luke's Episcopal Hospital, PO Box 20345 C1000, 77030, Houston, TX, USA.
| | - James T Willerson
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 77030, Houston, TX, USA. .,Texas Heart Institute at St. Luke's Episcopal Hospital, PO Box 20345 C1000, 77030, Houston, TX, USA.
| | - Richard Af Dixon
- Texas Heart Institute at St. Luke's Episcopal Hospital, PO Box 20345 C1000, 77030, Houston, TX, USA.
| |
Collapse
|
36
|
Abstract
Loss of cadherin 1 (CDH1; also known as epithelial cadherin (E-cadherin)) is used for the diagnosis and prognosis of epithelial cancers. However, it should not be ignored that the superfamily of transmembrane cadherin proteins encompasses more than 100 members in humans, including other classical cadherins, numerous protocadherins and cadherin-related proteins. Elucidation of their roles in suppression versus initiation or progression of various tumour types is a young but fascinating field of molecular cancer research. These cadherins are very diverse in both structure and function, and their mutual interactions seem to influence biological responses in complex and versatile ways.
Collapse
Affiliation(s)
- Frans van Roy
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.The Inflammation Research Center, VIB, B-9052 Ghent, Belgium
| |
Collapse
|
37
|
Lipopolysaccharide induces the interactions of breast cancer and endothelial cells via activated monocytes. Cancer Lett 2013; 345:75-84. [PMID: 24333719 DOI: 10.1016/j.canlet.2013.11.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 10/24/2013] [Accepted: 11/23/2013] [Indexed: 12/20/2022]
Abstract
The adhesion of circulating cancer cells to vascular endothelium is a key step in hematogenous metastasis. Cancer cell-endothelium interactions are mediated by cell adhesion molecules that can also be involved in the arrest of monocytes and other circulating leukocytes on endothelium in inflammation. Static and microfluidic flow adhesion assays as well as flow cytometry were conducted in this study to elucidate the role of monocytes, bacterial lipopolysaccharide (LPS), and histamine in breast cancer cell adhesion to vascular endothelial cells. Tumor necrosis factor-α (TNF-α) released from LPS-treated monocytes triggered the expression of intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) on endothelial cells. Histamine augmented the TNF-α effect, leading to a high number of arrested breast cancer cells under both static and shear flow conditions. LPS-treated monocytes were shown to enhance the arrest of breast cancer cells by anchoring the cancer cells to activated endothelial cells. This anchorage was achieved by binding cancer cell ICAM-1 to monocyte β2 integrins and binding endothelial ICAM-1 and VCAM-1 to monocyte β1 and β2 integrins. The results of this study imply that LPS is an important risk factor for cancer metastasis and that the elevated serum level of histamine further increases the risk of LPS-induced cancer metastasis. Preventing bacterial infections is essential in cancer treatment, and it is particularly vital for cancer patients affected by allergy.
Collapse
|
38
|
Haidari M, Zhang W, Wakame K. Disruption of endothelial adherens junction by invasive breast cancer cells is mediated by reactive oxygen species and is attenuated by AHCC. Life Sci 2013; 93:994-1003. [DOI: 10.1016/j.lfs.2013.10.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/16/2013] [Accepted: 10/25/2013] [Indexed: 11/30/2022]
|
39
|
Roh-Johnson M, Bravo-Cordero JJ, Patsialou A, Sharma VP, Guo P, Liu H, Hodgson L, Condeelis J. Macrophage contact induces RhoA GTPase signaling to trigger tumor cell intravasation. Oncogene 2013; 33:4203-12. [PMID: 24056963 PMCID: PMC3962803 DOI: 10.1038/onc.2013.377] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/01/2013] [Accepted: 07/16/2013] [Indexed: 12/23/2022]
Abstract
Most cancer patients die as a result of metastasis, thus it is important to understand the molecular mechanisms of dissemination, including intra- and extravasation. Although the mechanisms of extravasation have been vastly studied in vitro and in vivo, the process of intravasation is still unclear. Furthermore, how cells in the tumor microenvironment facilitate tumor cell intravasation is still unknown. Using high-resolution imaging, we found that macrophages enhance tumor cell intravasation upon physical contact. Macrophage and tumor cell contact induce RhoA activity in tumor cells, triggering the formation of actin-rich degradative protrusions called invadopodia, enabling tumor cells to degrade and break through matrix barriers during tumor cell transendothelial migration. Interestingly, we show that macrophage-induced invadopodium formation and tumor cell intravasation also occur in patient-derived tumor cells and in vivo models, revealing a conserved mechanism of tumor cell intravasation. Our results illustrate a novel heterotypic cell contact mediated signaling role for RhoA, as well as yield mechanistic insight into the ability of cells within the tumor microenvironment to facilitate steps of the metastatic cascade.
Collapse
Affiliation(s)
- M Roh-Johnson
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss-Lipper Biophotonics Center, Bronx, NY, USA
| | - J J Bravo-Cordero
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss-Lipper Biophotonics Center, Bronx, NY, USA
| | - A Patsialou
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss-Lipper Biophotonics Center, Bronx, NY, USA
| | - V P Sharma
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss-Lipper Biophotonics Center, Bronx, NY, USA
| | - P Guo
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss-Lipper Biophotonics Center, Bronx, NY, USA
| | - H Liu
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - L Hodgson
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss-Lipper Biophotonics Center, Bronx, NY, USA
| | - J Condeelis
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss-Lipper Biophotonics Center, Bronx, NY, USA
| |
Collapse
|
40
|
Fibrin and collagen differentially but synergistically regulate sprout angiogenesis of human dermal microvascular endothelial cells in 3-dimensional matrix. Int J Cell Biol 2013; 2013:231279. [PMID: 23737792 PMCID: PMC3657431 DOI: 10.1155/2013/231279] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 03/27/2013] [Accepted: 04/10/2013] [Indexed: 01/29/2023] Open
Abstract
Angiogenesis is a highly regulated event involving complex, dynamic interactions between microvascular endothelial cells and extracellular matrix (ECM) proteins. Alteration of ECM composition and architecture is a hallmark feature of wound clot and tumor stroma. We previously reported that during angiogenesis, endothelial cell responses to growth factors are modulated by the compositional and mechanical properties of a surrounding three-dimensional (3D) extracellular matrix (ECM) that is dominated by either cross-linked fibrin or type I collagen. However, the role of 3D ECM in the regulation of angiogenesis associated with wound healing and tumor growth is not well defined. This study investigates the correlation of sprout angiogenesis and ECM microenvironment using in vivo and in vitro 3D angiogenesis models. It demonstrates that fibrin and type I collagen 3D matrices differentially but synergistically regulate sprout angiogenesis. Thus blocking both integrin alpha v beta 3 and integrin alpha 2 beta 1 might be a novel strategy to synergistically block sprout angiogenesis in solid tumors.
Collapse
|
41
|
Evani SJ, Prabhu RG, Gnanaruban V, Finol EA, Ramasubramanian AK. Monocytes mediate metastatic breast tumor cell adhesion to endothelium under flow. FASEB J 2013; 27:3017-29. [PMID: 23616566 DOI: 10.1096/fj.12-224824] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Endothelial adhesion is necessary for the hematogenous dissemination of tumor cells. However, the metastatic breast tumor cell MDA-MB-231 does not bind to the endothelium under physiological flow conditions, suggesting alternate mechanisms of adhesion. Since monocytes are highly represented in the tumor microenvironment, and also bind to endothelium during inflammation, we hypothesized that the monocytes assist in the arrest of MDA-MB-231 on the endothelium. Using in vitro models of the dynamic shear environment of the vasculature, we show that TNF-α-activated THP1/primary human monocytes and MDA-MB-231 cells form stable aggregates, and that the monocytes in these aggregates mediate the adhesion of otherwise nonadherent MDA-MB-231 cells to inflamed endothelium under flow (55±2.4 vs. 1.7±0.82 at a shear stress of 0.5 dyn/cm(2), P<0.01). We also show that the hydrodynamic forces determine the size and orientation of aggregates adhered to the endothelium, and strongly favor the attachment of small aggregates with tumor cells downstream of flow (74-86% doublets at 0.5-2 dyn/cm(2), P<0.01). The 5-fold up-regulation of ICAM-1 on TNF-α-activated MDA-MB-231 cells through the Nf-κB pathway was found to be critical in MDA-MB-231-monocyte aggregation and endothelial adhesion. Our results demonstrate that, under inflammatory conditions, monocytes may serve to disseminate tumor cells through circulation, and the tumor-monocyte-endothelial axis may represent a new therapeutic target to reduce cancer metastasis.
Collapse
Affiliation(s)
- Shankar J Evani
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | | | | | | | | |
Collapse
|
42
|
Berdnikovs S, Abdala-Valencia H, Cook-Mills JM. Endothelial cell PTP1B regulates leukocyte recruitment during allergic inflammation. Am J Physiol Lung Cell Mol Physiol 2013; 304:L240-9. [PMID: 23275627 PMCID: PMC3567363 DOI: 10.1152/ajplung.00375.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 12/26/2012] [Indexed: 11/22/2022] Open
Abstract
Pulmonary eosinophilia is a consistent hallmark of allergic lung inflammation. Infiltration of eosinophils into ovalbumin (OVA)-challenged lungs is dependent on the adhesion molecule vascular cell adhesion molecule-1 (VCAM-1) on endothelial cells. Ligation of VCAM-1 activates endothelial cell protein tyrosine phosphatase 1B (PTP1B), which is required for VCAM-1-dependent leukocyte migration in vitro. To examine whether nonhematopoietic PTP1B modulates eosinophil recruitment in vivo, mice deficient in PTP1B were irradiated and received wild-type hematopoietic cells to generate chimeric PTP1B-/- mice. In response to OVA challenge, the chimeric PTP1B-/- mice had reduced eosinophilia in the lung tissue and bronchoalveolar lavage, indicating a role for PTP1B in nonhematopoietic cells during leukocyte recruitment. To determine whether endothelial cell PTP1B modulates eosinophil recruitment, mice with an inducible endothelial cell-specific PTP1B deletion (iePTP1B mice) were generated and the PTP1B deletion was induced after antigen sensitization before antigen challenge. In response to OVA challenge, the iePTP1B mice with the endothelial cell PTP1B deletion had an increased accumulation of eosinophils bound to the luminal surface of the endothelium in the lung vasculature and had a decrease in leukocyte recruitment into the lung tissue. In the iePTP1B mice, expression of adhesion molecules, cytokines, or chemokines that regulate leukocyte recruitment during inflammation was not altered, consistent with other studies that deletion of endothelial adhesion molecule signals does not alter lung cytokines and chemokines. In summary, these data suggest that VCAM-1 activation of PTP1B in the endothelium is necessary for eosinophil recruitment during allergic inflammation. Moreover, these studies provide a basis for targeting VCAM-1-dependent signaling pathways in allergy therapies.
Collapse
Affiliation(s)
- Sergejs Berdnikovs
- Division of Allergy and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | |
Collapse
|
43
|
Bouchard-Mercier A, Paradis AM, Rudkowska I, Lemieux S, Couture P, Vohl MC. Associations between dietary patterns and gene expression profiles of healthy men and women: a cross-sectional study. Nutr J 2013; 12:24. [PMID: 23398686 PMCID: PMC3598224 DOI: 10.1186/1475-2891-12-24] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 02/07/2013] [Indexed: 01/21/2023] Open
Abstract
Background Diet regulates gene expression profiles by several mechanisms. The objective of this study was to examine gene expression in relation with dietary patterns. Methods Two hundred and fifty four participants from the greater Quebec City metropolitan area were recruited. Two hundred and ten participants completed the study protocol. Dietary patterns were derived from a food frequency questionnaire (FFQ) by factor analysis. For 30 participants (in fasting state), RNA was extracted from peripheral blood mononuclear cells (PBMCs) and expression levels of 47,231 mRNA transcripts were assessed using the Illumina Human-6 v3 Expression BeadChips®. Microarray data was pre-processed with Flexarray software and analysed with Ingenuity Pathway Analysis (IPA). Results Two dietary patterns were identified. The Prudent dietary pattern was characterised by high intakes of vegetables, fruits, whole grain products and low intakes of refined grain products and the Western dietary pattern, by high intakes of refined grain products, desserts, sweets and processed meats. When individuals with high scores for the Prudent dietary pattern where compared to individuals with low scores, 2,083 transcripts were differentially expressed in men, 1,136 transcripts in women and 59 transcripts were overlapping in men and women. For the Western dietary pattern, 1,021 transcripts were differentially expressed in men with high versus low scores, 1,163 transcripts in women and 23 transcripts were overlapping in men and women. IPA reveals that genes differentially expressed for both patterns were present in networks related to the immune and/or inflammatory response, cancer and cardiovascular diseases. Conclusion Gene expression profiles were different according to dietary patterns, which probably modulate the risk of chronic diseases. Trial Registration NCT:
NCT01343342
Collapse
Affiliation(s)
- Annie Bouchard-Mercier
- Institute of Nutraceuticals and Functional Foods-INAF, Laval University, 2440 Hochelaga Blvd, Quebec G1V 0A6, Canada
| | | | | | | | | | | |
Collapse
|