201
|
Zheng M, Ambesi A, J. McKeown-Longo P. Role of TLR4 Receptor Complex in the Regulation of the Innate Immune Response by Fibronectin. Cells 2020; 9:cells9010216. [PMID: 31952223 PMCID: PMC7017243 DOI: 10.3390/cells9010216] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation and subsequent tissue fibrosis are associated with a biochemical and mechanical remodeling of the fibronectin matrix. Due to its conformational lability, fibronectin is considerably stretched by the contractile forces of the fibrotic microenvironment, resulting in the unfolding of its Type III domains. In earlier studies, we have shown that a peptide mimetic of a partially unfolded fibronectin Type III domain, FnIII-1c, functions as a Damage Associated Molecular Pattern (DAMP) molecule to induce activation of a toll-like receptor 4 (TLR4)/NF-B pathway and the subsequent release of fibro-inflammatory cytokines from human dermal fibroblasts. In the current study, we evaluated the requirement of the canonical TLR4/MD2/CD14 receptor complex in the regulation of FnIII-1c induced cytokine release. Using dermal fibroblasts and human embryonic kidney (HEK) cells, we found that all the components of the TLR4/MD2/CD14 complex were required for the release of the fibro-inflammatory cytokine, interleukin 8 (IL-8) in response to both FnIII-1c and the canonical TLR4 ligand, lipopolysaccharide (LPS). However, FnIII-1c mediated IL-8 release was strictly dependent on membrane-associated CD14, while LPS could use soluble CD14. These findings demonstrate that LPS and FnIII-1c share a similar but not identical mechanism of TLR4 activation in human dermal fibroblasts.
Collapse
|
202
|
Li Z, Shan X, Chen Z, Gao N, Zeng W, Zeng X, Mei L. Applications of Surface Modification Technologies in Nanomedicine for Deep Tumor Penetration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 8:2002589. [PMID: 33437580 PMCID: PMC7788636 DOI: 10.1002/advs.202002589] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/03/2020] [Indexed: 05/04/2023]
Abstract
The impermeable barrier of solid tumors due to the complexity of their components limits the treatment effect of nanomedicine and hinders its clinical translation. Several methods are available to increase the penetrability of nanomedicine, yet they are too complex to be effective, operational, or practical. Surface modification employs the characteristics of direct contact between multiphase surfaces to achieve the most direct and efficient penetration of solid tumors. Furthermore, their simple operation makes their use feasible. In this review, the latest surface modification strategies for the penetration of nanomedicine into solid tumors are summarized and classified into "bulldozer strategies" and "mouse strategies." Additionally, the evaluation methods, existing problems, and the development prospects of these technologies are discussed.
Collapse
Affiliation(s)
- Zimu Li
- Institute of PharmaceuticsSchool of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107China
| | - Xiaoting Shan
- Institute of PharmaceuticsSchool of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107China
| | - Zhidong Chen
- Institute of PharmaceuticsSchool of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107China
| | - Nansha Gao
- Institute of PharmaceuticsSchool of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107China
| | - Wenfeng Zeng
- Institute of PharmaceuticsSchool of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107China
| | - Xiaowei Zeng
- Institute of PharmaceuticsSchool of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107China
| | - Lin Mei
- Institute of PharmaceuticsSchool of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107China
- Tianjin Key Laboratory of Biomedical MaterialsKey Laboratory of Biomaterials and Nanotechnology for Cancer ImmunotherapyInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| |
Collapse
|
203
|
Wu Y, Ge G. Complexity of type IV collagens: from network assembly to function. Biol Chem 2019; 400:565-574. [PMID: 30864416 DOI: 10.1515/hsz-2018-0317] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023]
Abstract
Collagens form complex networks in the extracellular space that provide structural support and signaling cues to cells. Network-forming type IV collagens are the key structural components of basement membranes. In this review, we discuss how the complexity of type IV collagen networks is established, focusing on collagen α chain selection in type IV collagen protomer and network formation; covalent crosslinking in type IV collagen network stabilization; and the differences between solid-state type IV collagen in the extracellular matrix and soluble type IV collagen fragments. We further discuss how complex type IV collagen networks exert their physiological and pathological functions through cell surface integrin and nonintegrin receptors.
Collapse
Affiliation(s)
- Yuexin Wu
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Gaoxiang Ge
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
204
|
Khan FU, Owusu-Tieku NYG, Dai X, Liu K, Wu Y, Tsai HI, Chen H, Sun C, Huang L. Wnt/β-Catenin Pathway-Regulated Fibromodulin Expression Is Crucial for Breast Cancer Metastasis and Inhibited by Aspirin. Front Pharmacol 2019; 10:1308. [PMID: 31824307 PMCID: PMC6886402 DOI: 10.3389/fphar.2019.01308] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 10/15/2019] [Indexed: 01/08/2023] Open
Abstract
Emerging evidence suggests that fibromodulin (FMOD), an extracellular matrix protein, is associated with cancer, and yet little is known about the regulation of FMOD expression and its role in cancer metastasis. Aspirin, a classic anti-inflammatory drug, has been indicated to offer anticancer benefits, but its action targets and mechanisms remain obscure. In the present study using cell lines, animal model and database analysis, we show that FMOD is crucial for breast cancer cell migration and invasion (BCCMI) via activation of ERK; expression of FMOD is regulated positively by the Wnt/β-catenin pathway, wherein the β-catenin/TCF4/LEF1 complex binds the FMOD promoter to transcribe FMOD. Aspirin inhibits BCCMI by attenuating Wnt/β-catenin signaling and suppressing FMOD expression via inhibiting deacetylation of β-catenin by histone deacetylase 6 (HDAC6) leading to β-catenin phosphorylation and cytoplasmic degradation. Moreover, expression of the transcriptional complex components β-catenin/TCF4/LEF1 is upregulated by the Wnt/β-catenin pathway, constituting positive feedback loops that amplify its signal output. Our findings identify a critical role of FMOD in cancer metastasis, reveal a mechanism regulating FMOD transcription and impacting tumor metastasis, uncover action targets and mechanism for the anticancer activity of Aspirin, and expand the understanding of the Wnt/β-catenin pathway and tumor metastasis, which are valuable for development of cancer therapeutics.
Collapse
Affiliation(s)
- Fahim Ullah Khan
- School of Life Sciences, Tsinghua University, Beijing, China.,Shenzhen Key Laboratory of Gene and Antibody Therapy, Center for Biotechnology and Biomedicine, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology (prep), Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Nana Yaa Gyaama Owusu-Tieku
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen, China
| | - Xiaoyong Dai
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen, China
| | - Kewei Liu
- School of Life Sciences, Tsinghua University, Beijing, China.,Shenzhen Key Laboratory of Gene and Antibody Therapy, Center for Biotechnology and Biomedicine, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology (prep), Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Yanping Wu
- School of Life Sciences, Tsinghua University, Beijing, China.,Shenzhen Key Laboratory of Gene and Antibody Therapy, Center for Biotechnology and Biomedicine, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology (prep), Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Hsiang-I Tsai
- School of Life Sciences, Tsinghua University, Beijing, China.,Shenzhen Key Laboratory of Gene and Antibody Therapy, Center for Biotechnology and Biomedicine, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology (prep), Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Hongbo Chen
- School of Life Sciences, Tsinghua University, Beijing, China.,Shenzhen Key Laboratory of Gene and Antibody Therapy, Center for Biotechnology and Biomedicine, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology (prep), Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Chunhui Sun
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen, China
| | - Laiqiang Huang
- School of Life Sciences, Tsinghua University, Beijing, China.,Shenzhen Key Laboratory of Gene and Antibody Therapy, Center for Biotechnology and Biomedicine, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology (prep), Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China.,Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen, China
| |
Collapse
|
205
|
Lee JY, Dominguez AA, Nam S, Stowers RS, Qi LS, Chaudhuri O. Identification of cell context-dependent YAP-associated proteins reveals β 1 and β 4 integrin mediate YAP translocation independently of cell spreading. Sci Rep 2019; 9:17188. [PMID: 31748579 PMCID: PMC6868278 DOI: 10.1038/s41598-019-53659-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022] Open
Abstract
Yes-associated protein (YAP) is a transcriptional regulator and mechanotransducer, relaying extracellular matrix (ECM) stiffness into proliferative gene expression in 2D culture. Previous studies show that YAP activation is dependent on F-actin stress fiber mediated nuclear pore opening, however the protein mediators of YAP translocation remain unclear. Here, we show that YAP co-localizes with F-actin during activating conditions, such as sparse plating and culturing on stiff 2D substrates. To identify proteins mediating YAP translocation, we performed co-immunoprecipitation followed by mass spectrometry (co-IP/MS) for proteins that differentially associated with YAP under activating conditions. Interestingly, YAP preferentially associates with β1 integrin under activating conditions, and β4 integrin under inactivating conditions. In activating conditions, CRISPR/Cas9 knockout (KO) of β1 integrin (ΔITGB1) resulted in decreased cell area, which correlated with decreased YAP nuclear localization. ΔITGB1 did not significantly affect the slope of the correlation between YAP nuclear localization with area, but did decrease overall nuclear YAP independently of cell spreading. In contrast, β4 integrin KO (ΔITGB4) cells showed no change in cell area and similarly decreased nuclear YAP. These results reveal proteins that differentially associate with YAP during activation, which may aid in regulating YAP nuclear translocation.
Collapse
Affiliation(s)
- Joanna Y Lee
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, 94080, USA
| | - Antonia A Dominguez
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, 94305, USA
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Sungmin Nam
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Ryan S Stowers
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, 94305, USA
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
206
|
A Novel 3D Scaffold for Cell Growth to Asses Electroporation Efficacy. Cells 2019; 8:cells8111470. [PMID: 31752448 PMCID: PMC6912677 DOI: 10.3390/cells8111470] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/07/2019] [Accepted: 11/16/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor electroporation (EP) refers to the permeabilization of the cell membrane by means of short electric pulses thus allowing the potentiation of chemotherapeutic drugs. Standard plate adhesion 2D cell cultures can simulate the in vivo environment only partially due to lack of cell–cell interaction and extracellular matrix (ECM). In this study, we assessed a novel 3D scaffold for cell cultures based on hyaluronic acid and ionic-complementary self-assembling peptides (SAPs), by studying the growth patterns of two different breast carcinoma cell lines (HCC1569 and MDA-MB231). This 3D scaffold modulates cell shape and induces extracellular matrix deposit around cells. In the MDA-MB 231 cell line, it allows three-dimensional growth of structures known as spheroids, while in HCC1569 it achieves a cell organization similar to that observed in vivo. Interestingly, we were able to visualize the electroporation effect on the cells seeded in the new scaffold by means of standard propidium iodide assay and fluorescence microscopy. Thanks to the presence of cell–cell and cell–ECM interactions, the new 3D scaffold may represent a more reliable support for EP studies than 2D cancer cell cultures and may be used to test new EP-delivered drugs and novel EP protocols.
Collapse
|
207
|
Autenshlyus AI, Davletova KI, Studenikina AA, Mikhaylova ES, Varaksin NA, Zhurakovsky IP, Proskura AV, Sidorov SV, Lyakhovich VV. [Cytokine production by blood immune cells, tumor and its microenvironment, characteristic of extracellular matrix in patients with invasive ductal carcinoma of no special type]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:424-431. [PMID: 31666416 DOI: 10.18097/pbmc20196505424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The aim of this research was to study cytokine production by blood immune cells, tumor, and its microenvironment, and characterize extracellular matrix of patients with invasive ductal carcinoma of no special type and lymphatic metastases. Spontaneous and polyclonal activators stimulated production of cytokines by blood immune cells, tumor and its microenvironment were studied in 95 patients with invasive ductal carcinoma of no special type. The concentration of IL-2, IL-4, IL-6, IL-8, IL-10, IL-17, IL-18, IL-1β, IL-1Ra, TNF-α, IFN-γ, G-CSF, GM-CSF, VEGF and MCP-1 was determined by the solid-phase enzyme-linked immunosorbent assay. The condition of fibrous component and presence of neutral glycoproteins and sulfated glycosaminoglycans were evaluated during the research of extracellular matrix. Regional lymphatic metastases were detected in 35 of 95 patients. It was shown that in the presence or absence of lymphatic metastases index of polyclonal activators influence on the production of cytokines by blood immune cells was different for IL-6, IL-8, and IL-1β; while in the case of cytokine production by tumor and its microenvironment the index of influence was different for IL-2 and IL-17. The presence of lymphatic metastases corresponded with the rise of cytokines spontaneous production, while the absence of lymphatic metastases corresponded with the rise of cytokines production stimulated by polyclonal activators. The value of indices of polyclonal activators influence on the production of cytokines by blood immune cells pointed to the highly stimulating effect of polyclonal activators while the value of indices of polyclonal activators influence on cytokines production by tumor and its microenvironments pointed to the low and sometimes even absent effect of polyclonal activators. Basing on these data we propose a ratio of indices of polyclonal activators influence for the better evaluation of the probability of lymphatic metastases during preoperative period. After characterizing extracellular matrix we found out a point threshold, which, in 100% of cases, predicted the presence of lymphatic metastases basing on the condition of extracellular matrix. Using the data acquired, we are proposing a risk group for metastasis among women with no lymphatic metastases in the moment of check-up.
Collapse
Affiliation(s)
- A I Autenshlyus
- Novosibirsk State Medical University, Novosibirsk, Russia; Institute of Molecular Biology and Biophysics, Novosibirsk, Russia
| | - K I Davletova
- Novosibirsk State Medical University, Novosibirsk, Russia
| | | | - E S Mikhaylova
- Novosibirsk State Medical University, Novosibirsk, Russia; Institute of Molecular Biology and Biophysics, Novosibirsk, Russia
| | - N A Varaksin
- JSC "Vector-Best", Koltsovo, Novosibirsk Region, Russia
| | - I P Zhurakovsky
- Novosibirsk State Medical University, Novosibirsk, Russia; Institute of Molecular Biology and Biophysics, Novosibirsk, Russia
| | - A V Proskura
- Institute of Molecular Biology and Biophysics, Novosibirsk, Russia
| | - S V Sidorov
- Novosibirsk State University, Novosibirsk, Russia
| | - V V Lyakhovich
- Institute of Molecular Biology and Biophysics, Novosibirsk, Russia
| |
Collapse
|
208
|
MicroRNAs Contribute to Breast Cancer Invasiveness. Cells 2019; 8:cells8111361. [PMID: 31683635 PMCID: PMC6912645 DOI: 10.3390/cells8111361] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer statistics in 2018 highlight an 8.6 million incidence in female cancers, and 4.2 million cancer deaths globally. Moreover, breast cancer is the most frequent malignancy in females and twenty percent of these develop metastasis. This provides only a small chance for successful therapy, and identification of new molecular markers for the diagnosis and prognostic prediction of metastatic disease and development of innovative therapeutic molecules are therefore urgently required. Differentially expressed microRNAs (miRNAs) in cancers cause multiple changes in the expression of the tumorigenesis-promoting genes which have mostly been investigated in breast cancers. Herein, we summarize recent data on breast cancer-specific miRNA expression profiles and their participation in regulating invasive processes, in association with changes in cytoskeletal structure, cell-cell adhesion junctions, cancer cell-extracellular matrix interactions, tumor microenvironments, epithelial-to-mesenchymal transitions and cancer cell stem abilities. We then focused on the epigenetic regulation of individual miRNAs and their modified interactions with other regulatory genes, and reviewed the function of miRNA isoforms and exosome-mediated miRNA transfer in cancer invasiveness. Although research into miRNA’s function in cancer is still ongoing, results herein contribute to improved metastatic cancer management.
Collapse
|
209
|
Razazan A, Behravan J. Single peptides and combination modalities for triple negative breast cancer. J Cell Physiol 2019; 235:4089-4108. [PMID: 31642059 DOI: 10.1002/jcp.29300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/27/2019] [Indexed: 12/31/2022]
Abstract
Unlike other types of breast cancers (BCs), no specific therapeutic targets have been established for triple negative breast cancer (TNBC). Therefore, chemotherapy and radiotherapy are the only available adjuvant therapeutic choices for TNBC. New emerging reports show that TNBC is associated with higher numbers of intratumoral tumor infiltrating lymphocytes. This is indicative of host anti-TNBC immune surveillance and suggesting that immunotherapy can be considered as a therapeutic approach for TNBC management. Recent progress in molecular mechanisms of tumor-immune system interaction and cancer vaccine development studies, fast discoveries and FDA approvals of immune checkpoint inhibitors, chimeric antigen receptor T-cells, and oncolytic virotherapy have significantly attracted attention and research directions toward the immunotherapeutic approach to TNBC. Here in this review different aspects of TNBC immunotherapies including the host immune system-tumor interactions, the tumor microenvironment, the relevant molecular targets for immunotherapy, and clinical trials in the field are discussed.
Collapse
Affiliation(s)
- Atefeh Razazan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Center for Obesity, Diabetes and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, University of Waterloo, Waterloo, Canada.,Theraphage Inc., Kitchener, Ontario, Canada
| |
Collapse
|
210
|
Bray LJ, Hutmacher DW, Bock N. Addressing Patient Specificity in the Engineering of Tumor Models. Front Bioeng Biotechnol 2019; 7:217. [PMID: 31572718 PMCID: PMC6751285 DOI: 10.3389/fbioe.2019.00217] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer treatment is challenged by the heterogeneous nature of cancer, where prognosis depends on tumor type and disease stage, as well as previous treatments. Optimal patient stratification is critical for the development and validation of effective treatments, yet pre-clinical model systems are lacking in the delivery of effective individualized platforms that reflect distinct patient-specific clinical situations. Advances in cancer cell biology, biofabrication, and microengineering technologies have led to the development of more complex in vitro three-dimensional (3D) models to act as drug testing platforms and to elucidate novel cancer mechanisms. Mostly, these strategies have enabled researchers to account for the tumor microenvironment context including tumor-stroma interactions, a key factor of heterogeneity that affects both progression and therapeutic resistance. This is aided by state-of-the-art biomaterials and tissue engineering technologies, coupled with reproducible and high-throughput platforms that enable modeling of relevant physical and chemical factors. Yet, the translation of these models and technologies has been impaired by neglecting to incorporate patient-derived cells or tissues, and largely focusing on immortalized cell lines instead, contributing to drug failure rates. While this is a necessary step to establish and validate new models, a paradigm shift is needed to enable the systematic inclusion of patient-derived materials in the design and use of such models. In this review, we first present an overview of the components responsible for heterogeneity in different tumor microenvironments. Next, we introduce the state-of-the-art of current in vitro 3D cancer models employing patient-derived materials in traditional scaffold-free approaches, followed by novel bioengineered scaffold-based approaches, and further supported by dynamic systems such as bioreactors, microfluidics, and tumor-on-a-chip devices. We critically discuss the challenges and clinical prospects of models that have succeeded in providing clinical relevance and impact, and present emerging concepts of novel cancer model systems that are addressing patient specificity, the next frontier to be tackled by the field.
Collapse
Affiliation(s)
- Laura J. Bray
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Dietmar W. Hutmacher
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane, QLD, Australia
- Australian Research Council (ARC) Industrial Transformation Training Centre in Additive Biomanufacturing, Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane, QLD, Australia
| |
Collapse
|
211
|
Lee J, Park SH, Lee J, Chun H, Choi MK, Yoon JH, Pham TH, Kim KH, Kwon T, Ryu HW, Oh SR, Yoon DY. Differential effects of luteolin and its glycosides on invasion and apoptosis in MDA-MB-231 triple-negative breast cancer cells. EXCLI JOURNAL 2019; 18:750-763. [PMID: 31611756 PMCID: PMC6785773 DOI: 10.17179/excli2019-1459] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022]
Abstract
Luteolin is known to have anticancer activity in various cancers. Recent studies have shown that luteolin glycosides, such as luteolin-8-C-β-fucopyranoside, 7-methoxy-luteolin-8-C-β-(6- deoxyxylopyranos-3-uloside) and luteolin-8-C-β-d-glucopyranoside, flavonoids that are present in Arthraxon hispidus, exert antimigratory and anti-invasive effects, but no cytotoxic effect in estrogen receptor-positive MCF7 breast cancer cells. In the present study, we further investigated and compared differential effects of luteolin and its glycosides in MDA-MB-231 triple-negative breast cancer cells. Luteolin suppressed the expression of matrix metalloproteinase-9 and inhibited migration and invasion in MDA-MB-231 cells treated with the tumor promotor 12-O-tetradecanoylphorbol-13-acetate at non-cytotoxic concentrations (0, 5, and 10 μM). Furthermore, at cytotoxic concentrations (20 and 40 μM), luteolin induced apoptosis via extrinsic and intrinsic pathways in MDA-MB-231 cells. However, luteolin glycosides did not exert any cytotoxic, antimigratory, or anti-invasive effect in MDA-MB-231 cells. In brief, luteolin had both antimetastatic and cytotoxic effects on MDA-MB-231 cells, whereas luteolin glycosides had no effect on this cell line. Taking together the present results and our previous findings on the differential effects of luteolin and its glycosides on MDA-MB-231 and MCF-7 breast cancer cells, luteolin and its glycosides can be suggested as a potential candidate for breast cancer therapy.
Collapse
Affiliation(s)
- Jiyon Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Su-Ho Park
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jintak Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyunwoo Chun
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Myoung-Kwon Choi
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jae-Hwan Yoon
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Thu-Huyen Pham
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ki Hong Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Taeho Kwon
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyung-Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ohsong, Cheongju 28116, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ohsong, Cheongju 28116, Republic of Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
212
|
Tajbakhsh A, Rivandi M, Abedini S, Pasdar A, Sahebkar A. Regulators and mechanisms of anoikis in triple-negative breast cancer (TNBC): A review. Crit Rev Oncol Hematol 2019; 140:17-27. [DOI: 10.1016/j.critrevonc.2019.05.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 12/13/2018] [Accepted: 05/14/2019] [Indexed: 12/17/2022] Open
|
213
|
Bioinspired lipoproteins-mediated photothermia remodels tumor stroma to improve cancer cell accessibility of second nanoparticles. Nat Commun 2019; 10:3322. [PMID: 31346166 PMCID: PMC6658501 DOI: 10.1038/s41467-019-11235-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/26/2019] [Indexed: 02/07/2023] Open
Abstract
The tumor stromal microenvironments (TSM) including stromal cells and extracellular matrix (ECM) form an abominable barrier hampering nanoparticles accessibility to cancer cells, significantly compromising their antitumor effects. Herein, we report a bioinspired lipoprotein (bLP) that can induce efficient photothermia to remodel TSM and improve second bLP accessibility to cancer cells for antitumor therapy. The multiple stromal cells and ECM components in TSM are remarkably disrupted by bLP-mediated photothermal effects, which cause a 4.27-fold enhancement of second bLP accumulation in tumor, deep penetration in whole tumor mass and 27.0-fold increase of accessibility to cancer cells. Of note, this bLP-mediated TSM-remodeling to enhance cancer cell accessibility (TECA) strategy produces an eminent suppression of tumor growth and results in a 97.4% inhibition of lung metastasis, which is superior to the counterpart liposomes. The bLP-mediated TECA strategy provides deeper insights into enhancing nanoparticle accessibility to cancer cells for antitumor therapy. The stromal cells and extracellular matrix hamper nanoparticle access to cancer cells and their anti-cancer efficacy. Here, the authors report a bioinspired lipoprotein (bLP) for photothermal remodelling of tumour stroma and show this to improve subsequent bLP accessibility to cancer cells.
Collapse
|
214
|
Liu Y, Ye F. Construction and integrated analysis of crosstalking ceRNAs networks in laryngeal squamous cell carcinoma. PeerJ 2019; 7:e7380. [PMID: 31367490 PMCID: PMC6657684 DOI: 10.7717/peerj.7380] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/29/2019] [Indexed: 12/11/2022] Open
Abstract
Background Laryngeal squamous cell carcinoma (LSCC) is one of the most common malignant tumours of the head and neck. Recent evidence has demonstrated that lncRNAs play important roles in tumour progression and could be used as biomarkers for early diagnosis, prognosis, and potential therapeutic targets. The "competitive endogenous RNA (ceRNA)" hypothesis states that lncRNAs competitively bind to miRNAs through their intramolecular miRNA reaction elements (MREs) to construct a wide range of ceRNA regulatory networks. This study aims to predict the role of ceRNA network in LSCC, for advancing the understanding of underlying mechanisms of tumorigenesis. Material and Methods In this study, the functions of lncRNAs as ceRNAs in LSCC and their prognostic significance were investigated via comprehensive integrated expression profiles data of lncRNAs, mRNAs, and miRNAs obtained from The Cancer Genome Atlas (TCGA). Protein-protein interaction, gene ontology, pathway, and Kaplan-Meier curves analysis were used to profile the expression and function of altered RNAs in LSCC. Results As a result, 889 lncRNAs, 55 miRNAs and 1946 mRNAs were found to be differentially expressed in LSCC. These altered mRNAs were mainly involved in extracellular matrix organization, calcium signaling, and metabolic pathways. To study the regulatory function of lncRNAs, an lncRNA-mediated ceRNA network was constructed. This ceRNA network included 61 lncRNAs, seven miRNAs and seven target mRNAs. Of these RNAs, lncRNAs (TSPEAR-AS, CASK-AS1, MIR137HG, PART1, LSAMP-AS1), miRNA (has-mir-210) and mRNAs (HOXC13, STC2, DIO1, FOXD4L1) had a significant effect on the prognosis of LSCC. Conclusion The results of this study broaden the understanding of the mechanisms by which lncRNAs are involved in tumorigenesis. Furthermore, five lncRNAs (TSPEAR-AS, CASK-AS1, MIR137HG, PART1, LSAMP-AS1) were identified as potential prognostic biomarkers and therapeutic targets for LSCC. These results provide a basis for further experimental and clinical research.
Collapse
Affiliation(s)
- Yuehui Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Fan Ye
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| |
Collapse
|
215
|
Qi L, Gao C, Feng F, Zhang T, Yao Y, Wang X, Liu C, Li J, Li J, Sun C. MicroRNAs associated with lung squamous cell carcinoma: New prognostic biomarkers and therapeutic targets. J Cell Biochem 2019; 120:18956-18966. [PMID: 31241205 DOI: 10.1002/jcb.29216] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/31/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Lingyu Qi
- College of First Clinical MedicineShandong University of Traditional Chinese Medicine Jinan Shandong PR China
| | - Chundi Gao
- College of First Clinical MedicineShandong University of Traditional Chinese Medicine Jinan Shandong PR China
| | - Fubin Feng
- Department of OncologyWeifang Traditional Chinese Hospital Weifang Shandong PR China
| | - Tingting Zhang
- College of Traditional Chinese MedicineShandong University of Traditional Chinese Medicine Jinan Shandong PR China
| | - Yan Yao
- Clinical Medical CollegesWeifang Medical University Weifang Shandong PR China
| | - Xue Wang
- College of Basic MedicineQingdao University Qingdao Shandong PR China
| | - Cun Liu
- College of Traditional Chinese MedicineShandong University of Traditional Chinese Medicine Jinan Shandong PR China
| | - Jia Li
- Clinical Medical CollegesWeifang Medical University Weifang Shandong PR China
| | - Jie Li
- College of First Clinical MedicineShandong University of Traditional Chinese Medicine Jinan Shandong PR China
| | - Changgang Sun
- Department of OncologyAffiliated Hospital of Weifang Medical University Weifang Shandong PR China
- Department of OncologyAffiliated Hospital of Shandong University of Traditional Chinese Medicine Jinan Shandong PR China
| |
Collapse
|
216
|
Chen Z, Yan X, Li K, Ling Y, Kang H. Stromal fibroblast-derived MFAP5 promotes the invasion and migration of breast cancer cells via Notch1/slug signaling. Clin Transl Oncol 2019; 22:522-531. [PMID: 31190277 DOI: 10.1007/s12094-019-02156-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The tumor microenvironment (TME) regulates tumor progression, and cancer-associated fibroblasts (CAFs) are the primary stromal components of the TME, with the potential to drive tumor metastasis via the secretion of paracrine factors, but the specific mechanisms driving this process have not been defined. METHODS Proteins secreted from CAFs and normal fibroblasts (NFs) were analyzed via proteomic analysis (fold change > 2, p < 0.05) to identify tumor-promoting proteins secreted by CAFs. RESULTS Proteomic analysis revealed that microfibrillar-associated protein 5 (MFAP5) is preferentially expressed and secreted by CAFs relative to NFs, which was confirmed by Western blotting and RT-qPCR. Transwell and wound healing assays confirmed that MFAP5 is secreted by CAFs, and drives the invasion and migration of MCF7 breast cancer cells. We further found that in MCF7 cells MFAP5 promoted epithelial-mesenchymal transition, activating Notch1 signaling and consequently upregulating NICD1 and slug. When Notch1 was knocked down in MCF7 cells, the ability of MFAP5 to promote invasion and migration decreased. CONCLUSION CAFs promote cancer cells invasion and migration via MFAP5 secretion and activation of the Notch1/slug signaling. These data highlight this pathway as a therapeutic target to disrupt tumor progression through the interference of CAF-tumor crosstalk.
Collapse
Affiliation(s)
- Z Chen
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - X Yan
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - K Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Y Ling
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - H Kang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
217
|
Barupal DK, Gao B, Budczies J, Phinney BS, Perroud B, Denkert C, Fiehn O. Prioritization of metabolic genes as novel therapeutic targets in estrogen-receptor negative breast tumors using multi-omics data and text mining. Oncotarget 2019; 10:3894-3909. [PMID: 31231467 PMCID: PMC6570467 DOI: 10.18632/oncotarget.26995] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/13/2019] [Indexed: 01/12/2023] Open
Abstract
Estrogen-receptor negative (ERneg) breast cancer is an aggressive breast cancer subtype in the need for new therapeutic options. We have analyzed metabolomics, proteomics and transcriptomics data for a cohort of 276 breast tumors (MetaCancer study) and nine public transcriptomics datasets using univariate statistics, meta-analysis, Reactome pathway analysis, biochemical network mapping and text mining of metabolic genes. In the MetaCancer cohort, a total of 29% metabolites, 21% proteins and 33% transcripts were significantly different (raw p <0.05) between ERneg and ERpos breast tumors. In the nine public transcriptomics datasets, on average 23% of all genes were significantly different (raw p <0.05). Specifically, up to 60% of the metabolic genes were significantly different (meta-analysis raw p <0.05) across the transcriptomics datasets. Reactome pathway analysis of all omics showed that energy metabolism, and biosynthesis of nucleotides, amino acids, and lipids were associated with ERneg status. Text mining revealed that several significant metabolic genes and enzymes have been rarely reported to date, including PFKP, GART, PLOD1, ASS1, NUDT12, FAR1, PDE7A, FAHD1, ITPK1, SORD, HACD3, CDS2 and PDSS1. Metabolic processes associated with ERneg tumors were identified by multi-omics integration analysis of metabolomics, proteomics and transcriptomics data. Overall results suggested that TCA anaplerosis, proline biosynthesis, synthesis of complex lipids and mechanisms for recycling substrates were activated in ERneg tumors. Under-reported genes were revealed by text mining which may serve as novel candidates for drug targets in cancer therapies. The workflow presented here can also be used for other tumor types.
Collapse
Affiliation(s)
- Dinesh Kumar Barupal
- West Coast Metabolomics Center, University of California, Davis, CA, USA.,Co-first authors and contributed equally to this work
| | - Bei Gao
- West Coast Metabolomics Center, University of California, Davis, CA, USA.,Co-first authors and contributed equally to this work
| | - Jan Budczies
- Institute of Pathology, Charité University Hospital, Berlin, Germany
| | - Brett S Phinney
- UC Davis Genome Center, University of California, Davis, CA, USA
| | - Bertrand Perroud
- UC Davis Genome Center, University of California, Davis, CA, USA
| | - Carsten Denkert
- Institute of Pathology, Charité University Hospital, Berlin, Germany.,German Institute of Pathology, Philipps-University Marburg, Marburg, Germany
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA, USA
| |
Collapse
|
218
|
Insua-Rodríguez J, Pein M, Hongu T, Meier J, Descot A, Lowy CM, De Braekeleer E, Sinn HP, Spaich S, Sütterlin M, Schneeweiss A, Oskarsson T. Stress signaling in breast cancer cells induces matrix components that promote chemoresistant metastasis. EMBO Mol Med 2019; 10:emmm.201809003. [PMID: 30190333 PMCID: PMC6180299 DOI: 10.15252/emmm.201809003] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Metastatic progression remains a major burden for cancer patients and is associated with eventual resistance to prevailing therapies such as chemotherapy. Here, we reveal how chemotherapy induces an extracellular matrix (ECM), wound healing, and stem cell network in cancer cells via the c-Jun N-terminal kinase (JNK) pathway, leading to reduced therapeutic efficacy. We find that elevated JNK activity in cancer cells is linked to poor clinical outcome in breast cancer patients and is critical for tumor initiation and metastasis in xenograft mouse models of breast cancer. We show that JNK signaling enhances expression of the ECM and stem cell niche components osteopontin, also called secreted phosphoprotein 1 (SPP1), and tenascin C (TNC), that promote lung metastasis. We demonstrate that both SPP1 and TNC are direct targets of the c-Jun transcription factor. Exposure to multiple chemotherapies further exploits this JNK-mediated axis to confer treatment resistance. Importantly, JNK inhibition or disruption of SPP1 or TNC expression sensitizes experimental mammary tumors and metastases to chemotherapy, thus providing insights to consider for future treatment strategies against metastatic breast cancer.
Collapse
Affiliation(s)
- Jacob Insua-Rodríguez
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Maren Pein
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Tsunaki Hongu
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Physiological Chemistry and Department of Environmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Jasmin Meier
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Arnaud Descot
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Camille M Lowy
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Etienne De Braekeleer
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Peter Sinn
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Saskia Spaich
- Department of Obstetrics and Gynecology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Marc Sütterlin
- Department of Obstetrics and Gynecology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases-NCT, Heidelberg, Germany.,Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Thordur Oskarsson
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany .,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
219
|
Wang K, Wang G, Huang S, Luo A, Jing X, Li G, Zhou Y, Zhao X. Association between TIMP-2 gene polymorphism and breast cancer in Han Chinese women. BMC Cancer 2019; 19:446. [PMID: 31088428 PMCID: PMC6518501 DOI: 10.1186/s12885-019-5655-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/29/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND TIMP-2 gene plays an important role in the development of breast cancer. The present study was conducted to evaluate whether TIMP-2 gene polymorphisms are associated with breast cancer risk in a Han Chinese cohort. METHODS Six single nucleotide polymorphisms (SNPs) within the TIMP-2 gene in 571 breast cancer and 578 healthy control subjects were genotyped through the Agena MassARRAY. Logistic regression analysis was used to assess the influence of TIMP-2 polymorphisms on breast cancer. Functional annotation of TIMP-2 variants and TIMP-2 expression were analyzed by bioinformatics. RESULTS Bioinformatics analysis found that rs4789936 was likely to affect transcription factor binding, motifs, DNase footprint, and DNase peaks; and TIMP-2 was under-expressed in breast cancer, the risk allele of rs4789936 was associated with increased expression of TIMP-2 in peripheral blood samples. Importantly, individuals carrying TIMP-2 rs2277698 T allele have a 19% lower risk of breast cancer than individuals with allele C, providing protection (OR = 0.81, 95%CI = 0.67-0.99, p = 0.041). In the breast cancer patients with c-erb positive and PR positive, when the CC genotype was used as a reference, individuals carrying the TT genotype increased the risk of breast cancer. Haplotype analysis showed "TCC" was associated with a reduced risk of breast cancer (OR = 0.79, 95%CI = 0.63-0.97, p = 0.028). CONCLUSION Our study indicated that TIMP-2 rs2277698 was associated with breast cancer susceptibility.
Collapse
Affiliation(s)
- Kai Wang
- Department of Internal Medicine Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 West Yanta Road, Xi'an, 710061, Shaanxi, China.,The Second Department of Spleen and Stomach, The Hospital of Traditional Chinese Medicine of Shaanxi Province, Xi'an, 710063, Shaanxi, China
| | - Guanying Wang
- Department of Internal Medicine Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Shangke Huang
- Department of Internal Medicine Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Anqi Luo
- Department of Internal Medicine Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xin Jing
- Department of Internal Medicine Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Gang Li
- The Second Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yi Zhou
- The Center for Medical Imaging, The Hospital of Traditional Chinese Medicine of Shaanxi Province, Xi'an, 710063, Shaanxi, China
| | - Xinhan Zhao
- Department of Internal Medicine Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
220
|
Jézéquel P, Guette C, Lasla H, Gouraud W, Boissard A, Guérin‐Charbonnel C, Campone M. iTRAQ‐Based Quantitative Proteomic Analysis Strengthens Transcriptomic Subtyping of Triple‐Negative Breast Cancer Tumors. Proteomics 2019; 19:e1800484. [DOI: 10.1002/pmic.201800484] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/28/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Pascal Jézéquel
- Unité de Bioinfomique Institut de Cancérologie de l'Ouest Bd Jacques Monod 44805 Saint Herblain Cedex France
- Unité Mixte de Génomique du Cancer Institut de Cancérologie de l'Ouest ‐ René Gauducheau Bd Jacques Monod 44805 Saint Herblain Cedex France
- INSERM U1232 44007 Nantes Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
| | - Catherine Guette
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
- Institut de Cancérologie de l'Ouest—Paul Papin 49055 Angers Cedex France
| | - Hamza Lasla
- Unité de Bioinfomique Institut de Cancérologie de l'Ouest Bd Jacques Monod 44805 Saint Herblain Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
| | - Wilfried Gouraud
- Unité de Bioinfomique Institut de Cancérologie de l'Ouest Bd Jacques Monod 44805 Saint Herblain Cedex France
- Unité Mixte de Génomique du Cancer Institut de Cancérologie de l'Ouest ‐ René Gauducheau Bd Jacques Monod 44805 Saint Herblain Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
| | - Alice Boissard
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
- Institut de Cancérologie de l'Ouest—Paul Papin 49055 Angers Cedex France
| | - Catherine Guérin‐Charbonnel
- Unité de Bioinfomique Institut de Cancérologie de l'Ouest Bd Jacques Monod 44805 Saint Herblain Cedex France
- Unité Mixte de Génomique du Cancer Institut de Cancérologie de l'Ouest ‐ René Gauducheau Bd Jacques Monod 44805 Saint Herblain Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
| | - Mario Campone
- INSERM U1232 44007 Nantes Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
- Oncologie Médicale Institut de Cancérologie de l'Ouest—René Gauducheau 44805 Saint Herblain Cedex France
| |
Collapse
|
221
|
Houthuijzen JM, Jonkers J. Cancer-associated fibroblasts as key regulators of the breast cancer tumor microenvironment. Cancer Metastasis Rev 2019; 37:577-597. [PMID: 30465162 DOI: 10.1007/s10555-018-9768-3] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tumor cells exist in close proximity with non-malignant cells. Extensive and multilayered crosstalk between tumor cells and stromal cells tailors the tumor microenvironment (TME) to support survival, growth, and metastasis. Fibroblasts are one of the largest populations of non-malignant host cells that can be found within the TME of breast, pancreatic, and prostate tumors. Substantial scientific evidence has shown that these cancer-associated fibroblasts (CAFs) are not only associated with tumors by proximity but are also actively recruited to developing tumors where they can influence other cells of the TME as well as influencing tumor cell survival and metastasis. This review discusses the impact of CAFs on breast cancer biology and highlights their heterogeneity, origin and their role in tumor progression, ECM remodeling, therapy resistance, metastasis, and the challenges ahead of targeting CAFs to improve therapy response.
Collapse
Affiliation(s)
- J M Houthuijzen
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - J Jonkers
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
222
|
Ben-David-Naim M, Dagan A, Grad E, Aizik G, Nordling-David MM, Morss Clyne A, Granot Z, Golomb G. Targeted siRNA Nanoparticles for Mammary Carcinoma Therapy. Cancers (Basel) 2019; 11:E442. [PMID: 30934857 PMCID: PMC6521050 DOI: 10.3390/cancers11040442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Non-viral, polymeric-based, siRNA nanoparticles (NPs) have been proposed as promising gene delivery systems. Encapsulating siRNA in targeted NPs could confer improved biological stability, extended half-life, enhanced permeability, effective tumor accumulation, and therapy. In this work, a peptide derived from apolipoprotein B100 (ApoB-P), the protein moiety of low-density lipoprotein, was used to target siRNA-loaded PEGylated NPs to the extracellular matrix/proteoglycans (ECM/PGs) of a mammary carcinoma tumor. siRNA against osteopontin (siOPN), a protein involved in breast cancer development and progression, was encapsulated into PEGylated poly(d,l-lactic-co-glycolic acid) (PLGA) NPs using the double emulsion solvent diffusion technique. The NPs obtained possessed desired physicochemical properties including ~200 nm size, a neutral surface charge, and high siOPN loading of ~5 µg/mg. ApoB-P-targeted NPs exhibited both enhanced binding to isolated ECM and internalization by MDA-MB-231 human mammary carcinoma cells, in comparison to non-targeted NPs. Increased accumulation of the targeted NPs was achieved in the primary mammary tumor of mice xenografted with MDA-MB-231 mammary carcinoma cells as well as in the lungs, one of the main sites affected by metastases. siOPN NPs treatment resulted in significant inhibition of tumor growth (similar bioactivity of both formulations), accompanied with significant reduction of OPN mRNA levels (~40% knockdown of mRNA levels). We demonstrated that targeted NPs possessed enhanced tumor accumulation with increased therapeutic potential in mice models of mammary carcinoma.
Collapse
Affiliation(s)
- Meital Ben-David-Naim
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Arie Dagan
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Etty Grad
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Gil Aizik
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Mirjam M Nordling-David
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Alisa Morss Clyne
- Department of Mechanical Engineering and Mechanics, Drexel University, Philadelphia, PA 19104, USA.
| | - Zvi Granot
- Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Gershon Golomb
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| |
Collapse
|
223
|
Kim JE, Reynolds DS, Zaman MH, Mak M. Characterization of the mechanical properties of cancer cells in 3D matrices in response to collagen concentration and cytoskeletal inhibitors. Integr Biol (Camb) 2019; 10:232-241. [PMID: 29620778 DOI: 10.1039/c8ib00044a] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cellular processes, such as cell migration, adhesion, and proliferation depend on the interaction between the intracellular environment and the extracellular matrix (ECM). While many studies have explored the role of the microenvironment on cell behavior, the influence of 3D matrix mechanics on intracellular activity is not completely understood. To characterize the relationship between the mechanical components of the microenvironment and intracellular behavior, we use particle-tracking microrheology of metastatic breast cancer cells embedded in 3D collagen gels to quantify the intracellular activity from which the molecular motor activity and stiffness can be determined. Our results show that increasing collagen concentration of the 3D environments leads to an increase in intracellular stiffness and motor activity. Furthermore, our studies demonstrate that intracellular fluctuations depend on collagen concentration, even in the presence of a number of frontline chemotherapeutic and anti-MMP drugs, indicating that ECM concentration is an important and indispensable parameter to consider in drug screening.
Collapse
Affiliation(s)
- Jessica E Kim
- Biomedical Engineering Department, Boston University, Boston, Massachusetts, USA.
| | | | | | | |
Collapse
|
224
|
Connexin 43 Loss Triggers Cell Cycle Entry and Invasion in Non-Neoplastic Breast Epithelium: A Role for Noncanonical Wnt Signaling. Cancers (Basel) 2019; 11:cancers11030339. [PMID: 30857262 PMCID: PMC6468895 DOI: 10.3390/cancers11030339] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/15/2019] [Accepted: 03/04/2019] [Indexed: 12/26/2022] Open
Abstract
(1) Background: The expression of connexin 43 (Cx43) is disrupted in breast cancer, and re-expression of this protein in human breast cancer cell lines leads to decreased proliferation and invasiveness, suggesting a tumor suppressive role. This study aims to investigate the role of Cx43 in proliferation and invasion starting from non-neoplastic breast epithelium. (2) Methods: Nontumorigenic human mammary epithelial HMT-3522 S1 cells and Cx43 shRNA-transfected counterparts were cultured under 2-dimensional (2-D) and 3-D conditions. (3) Results: Silencing Cx43 induced mislocalization of β-catenin and Scrib from apicolateral membrane domains in glandular structures or acini formed in 3-D culture, suggesting the loss of apical polarity. Cell cycle entry and proliferation were enhanced, concomitantly with c-Myc and cyclin D1 upregulation, while no detectable activation of Wnt/β-catenin signaling was observed. Motility and invasion were also triggered and were associated with altered acinar morphology and activation of ERK1/2 and Rho GTPase signaling, which acts downstream of the noncanonical Wnt pathway. The invasion of Cx43-shRNA S1 cells was observed only under permissive stiffness of the extracellular matrix (ECM). (4) Conclusion: Our results suggest that Cx43 controls proliferation and invasion in the normal mammary epithelium in part by regulating noncanonical Wnt signaling.
Collapse
|
225
|
Fostok SF, El-Sibai M, El-Sabban M, Talhouk RS. Gap Junctions and Wnt Signaling in the Mammary Gland: a Cross-Talk? J Mammary Gland Biol Neoplasia 2019; 24:17-38. [PMID: 30194659 DOI: 10.1007/s10911-018-9411-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/03/2018] [Indexed: 12/21/2022] Open
Abstract
Connexins (Cxs), the building blocks of gap junctions (GJs), exhibit spatiotemporal patterns of expression and regulate the development and differentiation of the mammary gland, acting via channel-dependent and channel-independent mechanisms. Impaired Cx expression and localization are reported in breast cancer, suggesting a tumor suppressive role for Cxs. The signaling events that mediate the role of GJs in the development and tumorigenesis of the mammary gland remain poorly identified. The Wnt pathways, encompassing the canonical or the Wnt/β-catenin pathway and the noncanonical β-catenin-independent pathway, also play important roles in those processes. Indeed, aberrant Wnt signaling is associated with breast cancer. Despite the coincident roles of Cxs and Wnt pathways, the cross-talk in the breast tissue is poorly defined, although this is reported in a number of other tissues. Our previous studies revealed a channel-independent role for Cx43 in inducing differentiation or suppressing tumorigenesis of mammary epithelial cells by acting as a negative regulator of the Wnt/β-catenin pathway. Here, we provide a brief overview of mammary gland development, with emphasis on the role of Cxs in development and tumorigenesis of this tissue. We also discuss the role of Wnt signaling in similar contexts, and review the literature illustrating interplay between Cxs and Wnt pathways.
Collapse
Affiliation(s)
- Sabreen F Fostok
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), P.O. Box: 11-0236, Beirut, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut (AUB), Beirut, Lebanon
| | - Rabih S Talhouk
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), P.O. Box: 11-0236, Beirut, Lebanon.
| |
Collapse
|
226
|
Raic A, Naolou T, Mohra A, Chatterjee C, Lee-Thedieck C. 3D models of the bone marrow in health and disease: yesterday, today and tomorrow. MRS COMMUNICATIONS 2019; 9:37-52. [PMID: 30931174 PMCID: PMC6436722 DOI: 10.1557/mrc.2018.203] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 09/10/2018] [Indexed: 05/12/2023]
Abstract
The complex interaction between hematopoietic stem cells (HSCs) and their microenvironment in the human bone marrow ensures a life-long blood production by balancing stem cell maintenance and differentiation. This so-called HSC niche can be disturbed by malignant diseases. Investigating their consequences on hematopoiesis requires deep understanding of how the niches function in health and disease. To facilitate this, biomimetic models of the bone marrow are needed to analyse HSC maintenance and hematopoiesis under steady-state and diseased conditions. Here, 3D bone marrow models, their fabrication methods (including 3D bioprinting) and implementations recapturing bone marrow functions in health and diseases, are presented.
Collapse
Affiliation(s)
- Annamarija Raic
- Karlsruhe Institute of Technology (KIT), Institute of Functional
Interfaces, 76344 Eggenstein-Leopoldshafen, Germany
| | - Toufik Naolou
- Karlsruhe Institute of Technology (KIT), Institute of Functional
Interfaces, 76344 Eggenstein-Leopoldshafen, Germany
| | - Anna Mohra
- Karlsruhe Institute of Technology (KIT), Institute of Functional
Interfaces, 76344 Eggenstein-Leopoldshafen, Germany
| | - Chandralekha Chatterjee
- Karlsruhe Institute of Technology (KIT), Institute of Functional
Interfaces, 76344 Eggenstein-Leopoldshafen, Germany
| | - Cornelia Lee-Thedieck
- Karlsruhe Institute of Technology (KIT), Institute of Functional
Interfaces, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
227
|
Sun X, Zhuang B, Zhang M, Jiang H, Jin Y. Intratumorally Injected Photothermal Agent-Loaded Photodynamic Nanocarriers for Ablation of Orthotopic Melanoma and Breast Cancer. ACS Biomater Sci Eng 2019; 5:724-739. [DOI: 10.1021/acsbiomaterials.8b01111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Xiaodong Sun
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
- Institute of Pharmacy, Pharmaceutical College of Henan University, Jin Ming Avenue, Kaifeng 475004, China
| | - Bo Zhuang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Mengmeng Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
- Institute of Pharmacy, Pharmaceutical College of Henan University, Jin Ming Avenue, Kaifeng 475004, China
| | - Heliu Jiang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
- Institute of Pharmacy, Pharmaceutical College of Henan University, Jin Ming Avenue, Kaifeng 475004, China
| |
Collapse
|
228
|
Costa DF, Sarisozen C, Torchilin VP. Synthesis of Doxorubicin and miRNA Stimuli-Sensitive Conjugates for Combination Therapy. Methods Mol Biol 2019; 1974:99-109. [PMID: 31098998 DOI: 10.1007/978-1-4939-9220-1_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Recent advances in combination therapy by using chemotherapeutic drugs and small noncoding RNAs have highlighted the need for optimization of such agents to allow their carriage in a single delivery system. This protocol details the synthesis of a doxorubicin prodrug, where a NHS coupling reaction was used to sensitize the drug to the proteolytic activity of tumor microenvironments. The design of a lipid-modified miRNA by an S-S coupling reaction is also described. Modification of both, doxorubicin and miRNA, facilitated their simultaneous incorporation into mixed micelles for use in combination therapy against tumor cells.
Collapse
Affiliation(s)
- Daniel F Costa
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
- CAPES Foundation, Ministry of Education of Brazil, Brasília, DF, Brazil
| | - Can Sarisozen
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA.
| |
Collapse
|
229
|
Mechanisms of Matrix-Induced Chemoresistance of Breast Cancer Cells-Deciphering Novel Potential Targets for a Cell Sensitization. Cancers (Basel) 2018; 10:cancers10120495. [PMID: 30563275 PMCID: PMC6315379 DOI: 10.3390/cancers10120495] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/23/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022] Open
Abstract
Tumor cell binding to microenvironment components such as collagen type 1 (COL1) attenuates the sensitivity to cytotoxic drugs like cisplatin (CDDP) or mitoxantrone (MX), referred to as cell adhesion mediated drug resistance (CAM-DR). CAM-DR is considered as the onset for resistance mutations, but underlying mechanisms remain elusive. To evaluate CAM-DR as target for sensitization strategies, we analyzed signaling pathways in human estrogen-positive MCF-7 and triple-negative MDA-MB-231 breast cancer cells by western blot, proteome profiler array and TOP-flash assay in presence of COL1. β1-Integrins, known to bind COL1, appear as key for mediating COL1-related resistance in both cell lines that primarily follows FAK/PI3K/AKT pathway in MCF-7, and MAPK pathway in MDA-MB-231 cells. Notably, pCREB is highly elevated in both cell lines. Consequently, blocking these pathways sensitizes the cells evidently to CDDP and MX treatment. Wnt signaling is not relevant in this context. A β1-integrin knockdown of MCF-7 cells (MCF-7-β1-kd) reveals a signaling shift from FAK/PI3K/AKT to MAPK pathway, thus CREB emerges as a promising primary target for sensitization in MDA-MB-231, and secondary target in MCF-7 cells. Concluding, we provide evidence for importance of CAM-DR in breast cancer cells and identify intracellular signaling pathways as targets to sensitize cells for cytotoxicity treatment regimes.
Collapse
|
230
|
Toraskar J, Magnussen SN, Chawla K, Svineng G, Steigedal TS. Nephronectin mediates p38 MAPK-induced cell viability via its integrin-binding enhancer motif. FEBS Open Bio 2018; 8:1992-2001. [PMID: 30524949 PMCID: PMC6275265 DOI: 10.1002/2211-5463.12544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/23/2018] [Accepted: 10/08/2018] [Indexed: 12/16/2022] Open
Abstract
Nephronectin (NPNT) is an extracellular matrix (ECM) protein involved in kidney development. We recently reported intracellular NPNT as a potential prognostic marker in breast cancer and that NPNT promotes metastasis in an integrin-dependent manner. Here, we used reverse-phase protein array (RPPA) to analyze NPNT-triggered intracellular signaling in the 66cl4 mouse breast cancer cell line. The results showed that the integrin-binding enhancer motif is important for the cellular effects upon NPNT interaction with its receptors, including phosphorylation of p38 mitogen-activated protein kinase (MAPK). Furthermore, analysis using prediction tools suggests involvement of NPNT in promoting cell viability. In conclusion, our results indicate that NPNT, via its integrin-binding motifs, promotes cell viability through phosphorylation of p38 MAPK.
Collapse
Affiliation(s)
- Jimita Toraskar
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
- Central Norway Regional Health AuthorityStjørdalNorway
| | - Synnøve N. Magnussen
- Department of Medical BiologyFaculty of Health SciencesUiT‐The Arctic University of NorwayTromsøNorway
| | - Konika Chawla
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
- Bioinformatics Core Facility‐BioCoreNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Gunbjørg Svineng
- Department of Medical BiologyFaculty of Health SciencesUiT‐The Arctic University of NorwayTromsøNorway
| | - Tonje S. Steigedal
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
- Central Norway Regional Health AuthorityStjørdalNorway
| |
Collapse
|
231
|
Natividad RJ, Lalli ML, Muthuswamy SK, Asthagiri AR. Golgi Stabilization, Not Its Front-Rear Bias, Is Associated with EMT-Enhanced Fibrillar Migration. Biophys J 2018; 115:2067-2077. [PMID: 30366626 PMCID: PMC6343588 DOI: 10.1016/j.bpj.2018.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/24/2018] [Accepted: 10/05/2018] [Indexed: 01/06/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) and maturation of collagen fibrils in the tumor microenvironment play a significant role in cancer cell invasion and metastasis. Confinement along fiber-like tracks enhances cell migration. To what extent and in what manner EMT further promotes migration in a microenvironment already conducive to migration is poorly understood. Here, we show that TGFβ-mediated EMT significantly enhances migration on fiber-like micropatterned tracks of collagen, doubling migration speed and tripling persistence relative to untreated mammary epithelial cells. Thus, cell-intrinsic EMT and extrinsic fibrillar tracks have nonredundant effects on motility. To better understand EMT-enhanced fibrillar migration, we investigated the regulation of Golgi positioning, which is involved in front-rear polarization and persistent cell migration. Confinement along fiber-like tracks has been reported to favor posterior Golgi positioning, whereas anterior positioning is observed during 2-day wound healing. Although EMT also regulates cell polarity, little is known about its effect on Golgi positioning. Here, we show that EMT induces a 2:1 rearward bias in Golgi positioning; however, positional bias explains less than 2% of single-cell variability in migration speed and persistence. Meanwhile, EMT significantly stabilizes Golgi positioning. Cells that enhance migration in response to TGFβ maintain Golgi position for 2- to 4-fold longer than nonresponsive counterparts irrespective of whether the Golgi is ahead or behind the nucleus. In fact, 28% of TGFβ-responsive cells exhibit a fully committed Golgi phenotype with the organelle either in the anterior or posterior position for over 90% of the time. Furthermore, single-cell differences in Golgi stability capture up to 18% of variations in migration speed. These results suggest a hypothesis that the Golgi may be part of a core physical scaffold that affects how cell-generated forces are distributed during migration. A stable scaffold would be expected to more consistently and therefore more productively distribute forces over time, leading to efficient migration.
Collapse
Affiliation(s)
- Robert J Natividad
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Mark L Lalli
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Senthil K Muthuswamy
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Anand R Asthagiri
- Department of Bioengineering, Northeastern University, Boston, Massachusetts; Department of Chemical Engineering, Northeastern University, Boston, Massachusetts; Department of Biology, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
232
|
Guo S, Deng CX. Effect of Stromal Cells in Tumor Microenvironment on Metastasis Initiation. Int J Biol Sci 2018; 14:2083-2093. [PMID: 30585271 PMCID: PMC6299363 DOI: 10.7150/ijbs.25720] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/02/2018] [Indexed: 12/11/2022] Open
Abstract
The cellular environment where tumor cells reside is called the tumor microenvironment (TME), which consists of borders, blood vessels, lymph vessels, extracellular matrix (ECM), stromal cells, immune/inflammatory cells, secreted proteins, RNAs and small organelles. By dynamically interacting with tumor cells, stromal cells participate in all stages of tumor initiation, progression, metastasis, recurrence and drug response, and consequently, affect the fate of patients. During the processes of tumor evolution and metastasis initiation, stromal cells in TME also experience some changes and play roles in both the suppression and promotion of metastasis, while the overall function of stromal cells is beneficial for cancer cell survival and movement. In this review, we examine the effects of stromal cells in TME on metastasis initiation, including angiogenesis, epithelial-mesenchymal transition (EMT) and invasion. We also highlight functions of proteins, RNAs and small organelles secreted by stromal cells in their influences on multiple stages of tumor metastasis.
Collapse
Affiliation(s)
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
233
|
Second-harmonic patterned polarization-analyzed reflection confocal microscopy of stromal collagen in benign and malignant breast tissues. Sci Rep 2018; 8:16243. [PMID: 30389994 PMCID: PMC6214917 DOI: 10.1038/s41598-018-34693-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 10/23/2018] [Indexed: 01/30/2023] Open
Abstract
We present the results of polarimetric analysis of collagen on varying pathologies of breast tissues using second-harmonic patterned polarization-analyzed reflection confocal (SPPARC) microscopy. Experiments are conducted on a breast tissue microarray having benign tissues (BT), malignant invasive lobular carcinoma (ILC), and benign stroma adjacent to the malignant tissues (called the benign adjacent tissue, or BAT). Stroma in BAT and ILC exhibit the largest parameter differences. We observe that stromal collagen readings in ILC show lower depolarization, lower diattenuation and higher linear degree-of-polarization values than stromal collagen in BAT. This suggests that the optical properties of collagen change most in the vicinity of tumors. A similar trend is also exhibited in the non-collagenous extrafibrillar matrix plus cells (EFMC) region. The three highlighted parameters show greatest sensitivity to changes in the polarization response of collagen between pathologies.
Collapse
|
234
|
Jin Q, Liu G, Li S, Yuan H, Yun Z, Zhang W, Zhang S, Dai Y, Ma Y. Decellularized breast matrix as bioactive microenvironment for in vitro three‐dimensional cancer culture. J Cell Physiol 2018; 234:3425-3435. [PMID: 30387128 DOI: 10.1002/jcp.26782] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 04/23/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Qin Jin
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock College of Life Science, Inner Mongolia University Hohhot Inner Mongolia China
- Department of Pathology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Gang Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock College of Life Science, Inner Mongolia University Hohhot Inner Mongolia China
| | - Shubin Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock College of Life Science, Inner Mongolia University Hohhot Inner Mongolia China
| | - Haihan Yuan
- Department of Obstetrics People's Hospital of Beijing Daxing District Beijing China
| | - Zhizhong Yun
- Centre of Reproductive Medicine Inner Mongolia Hospital Hohhot Inner Mongolia China
| | - Wenqi Zhang
- College of Basic Medical Wanna Medical School Wuhu China
| | - Shu Zhang
- Department of Pathology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Yanfeng Dai
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock College of Life Science, Inner Mongolia University Hohhot Inner Mongolia China
| | - Yuzhen Ma
- Centre of Reproductive Medicine Inner Mongolia Hospital Hohhot Inner Mongolia China
| |
Collapse
|
235
|
Fibulin-2 is required for basement membrane integrity of mammary epithelium. Sci Rep 2018; 8:14139. [PMID: 30237579 PMCID: PMC6148073 DOI: 10.1038/s41598-018-32507-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/17/2018] [Indexed: 12/25/2022] Open
Abstract
Fibulin-2 (FBLN2) is a secreted extracellular matrix glycoprotein which has been associated with tissue development and remodelling. In the mouse mammary gland, FBLN2 can be detected during ductal morphogenesis in cap cells and myoepithelial cells at puberty and early pregnancy, respectively. In an attempt to assign its function, we knocked down Fbln2 in the mouse mammary epithelial cell line EpH4. FBLN2 reduction led to an increase in the size of spheroidal structures when compared to scrambled control shRNA-transduced cells plated on Matrigel matrix. This phenotype was associated with a disruption of the collagen IV sheath around the epithelial spheroids and downregulation of integrin β1, suggesting a role for FBLN2 in stabilizing the basement membrane (BM). In contrast to mice, in normal adult human breast tissue, FBLN2 was detected in ductal stroma, and in the interlobular stroma, but was not detectable within the lobular regions. In tissue sections of 65 breast cancers FBLN2 staining was lost around malignant cells with retained staining in the neighbouring histologically normal tissue margins. These results are consistent with a role of FBLN2 in mammary epithelial BM stability, and that its down-regulation in breast cancer is associated with loss of the BM and early invasion.
Collapse
|
236
|
Smithmyer ME, Deng CC, Cassel SE, LeValley PJ, Sumerlin BS, Kloxin AM. Self-healing boronic acid-based hydrogels for 3D co-cultures. ACS Macro Lett 2018; 7:1105-1110. [PMID: 32832198 PMCID: PMC7437986 DOI: 10.1021/acsmacrolett.8b00462] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Synthetic hydrogels have been widely adopted as well-defined matrices for three-dimensional (3D) cell culture, with increasing interest in systems that enable the co-culture of multiple cell types for probing both cell-matrix and cell-cell interactions in studies of tissue regeneration and disease. We hypothesized that the unique dynamic covalent chemistry of self-healing hydrogels could be harnessed for not only the encapsulation and culture of human cells but also the subsequent construction of layered hydrogels for 3D co-cultures. To test this, we formed hydrogels using boronic acid-functionalized polymers and demonstrated their self-healing in the presence of physiologically-relevant cell culture media. Two model human cell lines, MDA-MB-231 breast cancer cells and CCL151 pulmonary fibroblasts, were encapsulated within these dynamic materials, and good viability was observed over time. Finally, self-healing of cut hydrogel 'blocks' laden with these different cell types was used to create layered hydrogels for the generation of a dynamic co-culture system. This work demonstrates the utility of self-healing materials for multi-dimensional cultures and establishes approaches broadly useful for a variety of biological applications.
Collapse
Affiliation(s)
- Megan E. Smithmyer
- Department of Chemical and Biomolecular Engineering, Colburn Laboratory 150 Academy Street, University of Delaware, Newark, Delaware 19716, United States
| | - Christopher C. Deng
- George and Josephine Butler Polymer Research Laboratory, Center for Macromolecular Science and Engineering, Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, Florida 32611, United States
| | - Samantha E. Cassel
- Department of Chemical and Biomolecular Engineering, Colburn Laboratory 150 Academy Street, University of Delaware, Newark, Delaware 19716, United States
| | - Paige J. LeValley
- Department of Chemical and Biomolecular Engineering, Colburn Laboratory 150 Academy Street, University of Delaware, Newark, Delaware 19716, United States
| | - Brent S. Sumerlin
- George and Josephine Butler Polymer Research Laboratory, Center for Macromolecular Science and Engineering, Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, Florida 32611, United States
| | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, Colburn Laboratory 150 Academy Street, University of Delaware, Newark, Delaware 19716, United States
- Department of Materials Science and Engineering, DuPont Hall, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
237
|
Gui P, Ben-Neji M, Belozertseva E, Dalenc F, Franchet C, Gilhodes J, Labrousse A, Bellard E, Golzio M, Poincloux R, Maridonneau-Parini I, Le Cabec V. The Protease-Dependent Mesenchymal Migration of Tumor-Associated Macrophages as a Target in Cancer Immunotherapy. Cancer Immunol Res 2018; 6:1337-1351. [DOI: 10.1158/2326-6066.cir-17-0746] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 06/29/2018] [Accepted: 08/20/2018] [Indexed: 11/16/2022]
|
238
|
Ansardamavandi A, Tafazzoli-Shadpour M, Shokrgozar MA. Behavioral remodeling of normal and cancerous epithelial cell lines with differing invasion potential induced by substrate elastic modulus. Cell Adh Migr 2018; 12:472-488. [PMID: 29969940 PMCID: PMC6363025 DOI: 10.1080/19336918.2018.1475803] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/07/2018] [Indexed: 12/20/2022] Open
Abstract
The micro-environment of cancer cells in the body is mechanically stiffer than that of normal cells. We cultured three breast cell lines of MCF10A-normal, MCF7-noninvasive, and MDA-MB-231-invasive on PDMS substrates with different elastic moduli and different cellular features were examined.Effects of substrate stiffness on cell behavior were evident among all cell lines. Cancerous cells were more sensitive to substrate stiffness for cell behaviors related to cell motility and migration which are necessary for invasion. The invasive cancerous cells were the most motile on substrates with moderate stiffness followed by non-invasive cancerous cells. Gene markers alterations were generally according to the analyzed cell movement parameters. Results suggest that alterations in matrix stiffness may be related to cancer disease and progression.
Collapse
Affiliation(s)
- Arian Ansardamavandi
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | | | | |
Collapse
|
239
|
Targeting cancer stem cells and their niche: perspectives for future therapeutic targets and strategies. Semin Cancer Biol 2018; 53:139-155. [PMID: 30081228 DOI: 10.1016/j.semcancer.2018.08.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/30/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023]
Abstract
A small subpopulation of cells within the bulk of tumors share features with somatic stem cells, in that, they are capable of self-renewal, they differentiate, and are highly resistant to conventional therapy. These cells have been referred to as cancer stem cells (CSCs). Recent reports support the central importance of a cancer stem cell-like niche that appears to help foster the generation and maintenance of CSCs. In response to signals provided by this microenvironment, CSCs express the tumorigenic characteristics that can drive tumor metastasis by the induction of epithelial-mesenchymal-transition (EMT) that in turn fosters the migration and recolonization of the cells as secondary tumors within metastatic niches. We summarize here recent advances in cancer stem cell research including the characterization of their genetic and epigenetic features, metabolic specialities, and crosstalk with aging-associated processes. Potential strategies for targeting CSCs, and their niche, by regulating CSCs plasticity, or therapeutic sensitivity is discussed. Finally, it is hoped that new strategies and related therapeutic approaches as outlined here may help prevent the formation of the metastatic niche, as well as counter tumor progression and metastatic growth.
Collapse
|
240
|
Spill F, Bakal C, Mak M. Mechanical and Systems Biology of Cancer. Comput Struct Biotechnol J 2018; 16:237-245. [PMID: 30105089 PMCID: PMC6077126 DOI: 10.1016/j.csbj.2018.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/03/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022] Open
Abstract
Mechanics and biochemical signaling are both often deregulated in cancer, leading toincreased cell invasiveness, proliferation, and survival. The dynamics and interactions of cytoskeletal components control basic mechanical properties, such as cell tension, stiffness, and engagement with the extracellular environment, which can lead to extracellular matrix remodeling. Intracellular mechanics can alter signaling and transcription factors, impacting cell decision making. Additionally, signaling from soluble and mechanical factors in the extracellular environment, such as substrate stiffness and ligand density, can modulate cytoskeletal dynamics. Computational models closely integrated with experimental support, incorporating cancer-specific parameters, can provide quantitative assessments and serve as predictive tools toward dissecting the feedback between signaling and mechanics and across multiple scales and domains in tumor progression.
Collapse
Affiliation(s)
- Fabian Spill
- School of Mathematics, University of Birmingham, Birmingham B15 2TT, UK
| | - Chris Bakal
- Division of Cancer Biology, Chester Beatty Laboratories, The Institute of Cancer Research, London SW3 6JB, UK
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, USA
| |
Collapse
|
241
|
Socovich AM, Naba A. The cancer matrisome: From comprehensive characterization to biomarker discovery. Semin Cell Dev Biol 2018; 89:157-166. [PMID: 29964200 DOI: 10.1016/j.semcdb.2018.06.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/18/2018] [Accepted: 06/27/2018] [Indexed: 02/07/2023]
Abstract
Tumor progression and dissemination critically depend on support from the tumor microenvironment, the ensemble of cellular and acellular components surrounding and interacting with tumor cells. The extracellular matrix (ECM), the complex scaffolding of hundreds of proteins organizing cells in tissues, is a major component of the tumor microenvironment. It orchestrates cellular processes including proliferation, migration, and invasion, that are highly dysregulated during cancer progression. Alterations in ECM abundance, integrity, and mechanical properties have been correlated with poorer prognosis for cancer patients. Yet the ECM proteome, or "matrisome," of tumors remained until recently largely unexplored. This review will present the recent developments in computational and proteomic technologies that have allowed the comprehensive characterization of the ECM of different tumor types and microenvironmental niches. These approaches have resulted in the definition of protein signatures distinguishing tumors from normal tissues, tumors of different stages, primary from secondary tumors, and tumors from other diseased states such as fibrosis. Moreover, recent studies have demonstrated that the levels of expression of certain genes encoding ECM and ECM-associated proteins is prognostic of cancer patient survival and can thus serve as biomarkers. Last, proteomic studies have permitted the identification of novel ECM proteins playing functional roles in cancer progression. Such proteins have the potential to be exploited as therapeutic targets.
Collapse
Affiliation(s)
- Alexandra M Socovich
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
242
|
Chen Y, Tan W, Wang C. Tumor-associated macrophage-derived cytokines enhance cancer stem-like characteristics through epithelial-mesenchymal transition. Onco Targets Ther 2018; 11:3817-3826. [PMID: 30013362 PMCID: PMC6038883 DOI: 10.2147/ott.s168317] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cells are a small population of cells with the potential for self-renewal and multi-directional differentiation and are an important source of cancer initiation, treatment resistance, and recurrence. Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells lose their epithelial phenotype and convert to mesenchymal cells. Recent studies have shown that cancer cells undergoing EMT can become stem-like cells. Many kinds of tumors are associated with chronic inflammation, which plays a role in tumor progression. Among the various immune cells mediating chronic inflammation, macrophages account for ~30%-50% of the tumor mass. Macrophages are highly infiltrative in the tumor microenvironment and secrete a series of inflammatory factors and cytokines, such as transforming growth factor (TGF)-β, IL-6, IL-10, and tumor necrosis factor (TNF)-α, which promote EMT and enhance the stemness of cancer cells. This review summarizes and discusses recent research findings on some specific mechanisms of tumor-associated macrophage-derived cytokines in EMT and cancer stemness transition, which are emerging targets of cancer treatment.
Collapse
Affiliation(s)
- Yongxu Chen
- Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, Guangdong Province, People's Republic of China, .,School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, People's Republic of China,
| | - Wei Tan
- Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, Guangdong Province, People's Republic of China,
| | - Changjun Wang
- Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, Guangdong Province, People's Republic of China, .,School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, People's Republic of China,
| |
Collapse
|
243
|
Fiorino S, Di Saverio S, Leandri P, Tura A, Birtolo C, Silingardi M, de Biase D, Avisar E. The role of matricellular proteins and tissue stiffness in breast cancer: a systematic review. Future Oncol 2018; 14:1601-1627. [PMID: 29939077 DOI: 10.2217/fon-2017-0510] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Malignancies consist not only of cancerous and nonmalignant cells, but also of additional elements, as extracellular matrix. The aim of this review is to summarize meta-analyses, describing breast tissue stiffness and risk of breast carcinoma (BC) assessing the potential relationship between matricellular proteins (MPs) and survival. A systematic computer-based search of published articles, according to PRISMA statement, was conducted through Ovid interface. Mammographic density and tissue stiffness are associated with the risk of BC development, suggesting that MPs may influence BC prognosis. No definitive conclusions are available and additional researches are required to definitively clarify the role of each MP, mammographic density and stiffness in BC development and the mechanisms involved in the onset of this malignancy.
Collapse
Affiliation(s)
- Sirio Fiorino
- Internal Medicine 'C' Unit, Maggiore Hospital, Local Health Unit of Bologna, Bologna, Italy
| | - Salomone Di Saverio
- Cambridge Colorectal Unit, Box 201, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Paolo Leandri
- Internal Medicine 'C' Unit, Maggiore Hospital, Local Health Unit of Bologna, Bologna, Italy
| | - Andrea Tura
- Metabolic Unit, CNR Institute of Neuroscience, Padova, Italy
| | - Chiara Birtolo
- Geriatric Unit, Azienda USL-Maggiore Hospital, Largo Nigrisoli 3, Bologna, Italy
| | - Mauro Silingardi
- Internal Medicine 'A' Unit, Maggiore Hospital, Local Health Unit of Bologna, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy & Biotechnology, Molecular Pathology Unit, University of Bologna, Bologna, Italy
| | - Eli Avisar
- Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
244
|
Dower CM, Wills CA, Frisch SM, Wang HG. Mechanisms and context underlying the role of autophagy in cancer metastasis. Autophagy 2018; 14:1110-1128. [PMID: 29863947 DOI: 10.1080/15548627.2018.1450020] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Macroautophagy/autophagy is a fundamental cellular degradation mechanism that maintains cell homeostasis, regulates cell signaling, and promotes cell survival. Its role in promoting tumor cell survival in stress conditions is well characterized, and makes autophagy an attractive target for cancer therapy. Emerging research indicates that autophagy also influences cancer metastasis, which is the primary cause of cancer-associated mortality. However, data demonstrate that the regulatory role of autophagy in metastasis is multifaceted, and includes both metastasis-suppressing and -promoting functions. The metastasis-suppressing functions of autophagy, in particular, have important implications for autophagy-based treatments, as inhibition of autophagy may increase the risk of metastasis. In this review, we discuss the mechanisms and context underlying the role of autophagy in metastasis, which include autophagy-mediated regulation of focal adhesion dynamics, integrin signaling and trafficking, Rho GTPase-mediated cytoskeleton remodeling, anoikis resistance, extracellular matrix remodeling, epithelial-to-mesenchymal transition signaling, and tumor-stromal cell interactions. Through this, we aim to clarify the context-dependent nature of autophagy-mediated metastasis and provide direction for further research investigating the role of autophagy in cancer metastasis.
Collapse
Affiliation(s)
- Christopher M Dower
- a Department of Pediatrics , Pennsylvania State University College of Medicine , Hershey , PA USA
| | - Carson A Wills
- a Department of Pediatrics , Pennsylvania State University College of Medicine , Hershey , PA USA
| | - Steven M Frisch
- b WVU Cancer Institute, Department of Biochemistry , West Virginia University , Morgantown , WV USA
| | - Hong-Gang Wang
- a Department of Pediatrics , Pennsylvania State University College of Medicine , Hershey , PA USA
| |
Collapse
|
245
|
Belgodere JA, King CT, Bursavich JB, Burow ME, Martin EC, Jung JP. Engineering Breast Cancer Microenvironments and 3D Bioprinting. Front Bioeng Biotechnol 2018; 6:66. [PMID: 29881724 PMCID: PMC5978274 DOI: 10.3389/fbioe.2018.00066] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a critical cue to direct tumorigenesis and metastasis. Although two-dimensional (2D) culture models have been widely employed to understand breast cancer microenvironments over the past several decades, the 2D models still exhibit limited success. Overwhelming evidence supports that three dimensional (3D), physiologically relevant culture models are required to better understand cancer progression and develop more effective treatment. Such platforms should include cancer-specific architectures, relevant physicochemical signals, stromal-cancer cell interactions, immune components, vascular components, and cell-ECM interactions found in patient tumors. This review briefly summarizes how cancer microenvironments (stromal component, cell-ECM interactions, and molecular modulators) are defined and what emerging technologies (perfusable scaffold, tumor stiffness, supporting cells within tumors and complex patterning) can be utilized to better mimic native-like breast cancer microenvironments. Furthermore, this review emphasizes biophysical properties that differ between primary tumor ECM and tissue sites of metastatic lesions with a focus on matrix modulation of cancer stem cells, providing a rationale for investigation of underexplored ECM proteins that could alter patient prognosis. To engineer breast cancer microenvironments, we categorized technologies into two groups: (1) biochemical factors modulating breast cancer cell-ECM interactions and (2) 3D bioprinting methods and its applications to model breast cancer microenvironments. Biochemical factors include matrix-associated proteins, soluble factors, ECMs, and synthetic biomaterials. For the application of 3D bioprinting, we discuss the transition of 2D patterning to 3D scaffolding with various bioprinting technologies to implement biophysical cues to model breast cancer microenvironments.
Collapse
Affiliation(s)
- Jorge A. Belgodere
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Connor T. King
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Jacob B. Bursavich
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Matthew E. Burow
- Department of Medicine, Section Hematology/Oncology, Tulane University, New Orleans, LA, United States
| | - Elizabeth C. Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Jangwook P. Jung
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
246
|
Logun M, Zhao W, Mao L, Karumbaiah L. Microfluidics in Malignant Glioma Research and Precision Medicine. ADVANCED BIOSYSTEMS 2018; 2:1700221. [PMID: 29780878 PMCID: PMC5959050 DOI: 10.1002/adbi.201700221] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Indexed: 01/09/2023]
Abstract
Glioblastoma multiforme (GBM) is an aggressive form of brain cancer that has no effective treatments and a prognosis of only 12-15 months. Microfluidic technologies deliver microscale control of fluids and cells, and have aided cancer therapy as point-of-care devices for the diagnosis of breast and prostate cancers. However, a few microfluidic devices are developed to study malignant glioma. The ability of these platforms to accurately replicate the complex microenvironmental and extracellular conditions prevailing in the brain and facilitate the measurement of biological phenomena with high resolution and in a high-throughput manner could prove useful for studying glioma progression. These attributes, coupled with their relatively simple fabrication process, make them attractive for use as point-of-care diagnostic devices for detection and treatment of GBM. Here, the current issues that plague GBM research and treatment, as well as the current state of the art in glioma detection and therapy, are reviewed. Finally, opportunities are identified for implementing microfluidic technologies into research and diagnostics to facilitate the rapid detection and better therapeutic targeting of GBM.
Collapse
Affiliation(s)
- Meghan Logun
- Regenerative Bioscience Center, ADS Complex, University of Georgia, 425 River Road, Athens, GA 30602-2771, USA
| | - Wujun Zhao
- Department of Chemistry, University of Georgia, Athens, GA 30602-2771, USA
| | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA 30602-2771, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, ADS Complex, University of Georgia, 425 River Road, Athens, GA 30602-2771, USA
| |
Collapse
|
247
|
Jokela TA, Engelsen AST, Rybicka A, Pelissier Vatter FA, Garbe JC, Miyano M, Tiron C, Ferariu D, Akslen LA, Stampfer MR, Lorens JB, LaBarge MA. Microenvironment-Induced Non-sporadic Expression of the AXL and cKIT Receptors Are Related to Epithelial Plasticity and Drug Resistance. Front Cell Dev Biol 2018; 6:41. [PMID: 29719832 PMCID: PMC5913284 DOI: 10.3389/fcell.2018.00041] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/23/2018] [Indexed: 12/13/2022] Open
Abstract
The existence of rare cancer cells that sporadically acquire drug-tolerance through epigenetic mechanisms is proposed as one mechanism that drives cancer therapy failure. Here we provide evidence that specific microenvironments impose non-sporadic expression of proteins related to epithelial plasticity and drug resistance. Microarrays of robotically printed combinatorial microenvironments of known composition were used to make cell-based functional associations between microenvironments, which were design-inspired by normal and tumor-burdened breast tissues, and cell phenotypes. We hypothesized that specific combinations of microenvironment constituents non-sporadically impose the induction of the AXL and cKIT receptor tyrosine kinase proteins, which are known to be involved in epithelial plasticity and drug-tolerance, in an isogenic human mammary epithelial cell (HMEC) malignant progression series. Dimension reduction analysis reveals type I collagen as a dominant feature, inducing expression of both markers in pre-stasis finite lifespan HMECs, and transformed non-malignant and malignant immortal cell lines. Basement membrane-associated matrix proteins, laminin-111 and type IV collagen, suppress AXL and cKIT expression in pre-stasis and non-malignant cells. However, AXL and cKIT are not suppressed by laminin-111 in malignant cells. General linear models identified key factors, osteopontin, IL-8, and type VIα3 collagen, which significantly upregulated AXL and cKIT, as well as a plasticity-related gene expression program that is often observed in stem cells and in epithelial-to-mesenchymal-transition. These factors are co-located with AXL-expressing cells in situ in normal and breast cancer tissues, and associated with resistance to paclitaxel. A greater diversity of microenvironments induced AXL and cKIT expression consistent with plasticity and drug-tolerant phenotypes in tumorigenic cells compared to normal or immortal cells, suggesting a reduced perception of microenvironment specificity in malignant cells. Microenvironment-imposed reprogramming could explain why resistant cells are seemingly persistent and rapidly adaptable to multiple classes of drugs. These results support the notion that specific microenvironments drive drug-tolerant cellular phenotypes and suggest a novel interventional avenue for preventing acquired therapy resistance.
Collapse
Affiliation(s)
- Tiina A. Jokela
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Population Sciences, Center for Cancer and Aging, City of Hope, Duarte, CA, United States
| | - Agnete S. T. Engelsen
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Agata Rybicka
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - James C. Garbe
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Masaru Miyano
- Department of Population Sciences, Center for Cancer and Aging, City of Hope, Duarte, CA, United States
| | - Crina Tiron
- Regional Institute of Oncology, Iasi, Romania
| | - Dan Ferariu
- Regional Institute of Oncology, Iasi, Romania
| | - Lars A. Akslen
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Martha R. Stampfer
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - James B. Lorens
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Mark A. LaBarge
- Department of Population Sciences, Center for Cancer and Aging, City of Hope, Duarte, CA, United States
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| |
Collapse
|
248
|
An overview of mammographic density and its association with breast cancer. Breast Cancer 2018; 25:259-267. [PMID: 29651637 PMCID: PMC5906528 DOI: 10.1007/s12282-018-0857-5] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/30/2018] [Indexed: 12/24/2022]
Abstract
In 2017, breast cancer became the most commonly diagnosed cancer among women in the US. After lung cancer, breast cancer is the leading cause of cancer-related mortality in women. The breast consists of several components, including milk storage glands, milk ducts made of epithelial cells, adipose tissue, and stromal tissue. Mammographic density (MD) is based on the proportion of stromal, epithelial, and adipose tissue. Women with high MD have more stromal and epithelial cells and less fatty adipose tissue, and are more likely to develop breast cancer in their lifetime compared to women with low MD. Because of this correlation, high MD is an independent risk factor for breast cancer. Further, mammographic screening is less effective in detecting suspicious lesions in dense breast tissue, which can lead to late-stage diagnosis. Molecular differences between dense and non-dense breast tissues explain the underlying biological reasons for why women with dense breasts are at a higher risk for developing breast cancer. The goal of this review is to highlight the current molecular understanding of MD, its association with breast cancer risk, the demographics pertaining to MD, and the environmental factors that modulate MD. Finally, we will review the current legislation regarding the disclosure of MD on a traditional screening mammogram and the supplemental screening options available to women with dense breast tissue.
Collapse
|
249
|
Xu M, Seas A, Kiyani M, Ji KSY, Bell HN. A temporal examination of calcium signaling in cancer- from tumorigenesis, to immune evasion, and metastasis. Cell Biosci 2018; 8:25. [PMID: 29636894 PMCID: PMC5883416 DOI: 10.1186/s13578-018-0223-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/26/2018] [Indexed: 12/16/2022] Open
Abstract
Background Although the study of calcium (Ca2+) is classically associated with excitable cells such as myocytes or neurons, the ubiquity of this essential element in all cellular processes has led to interest in other cell types. The importance of Ca2+ to apoptosis, cell signaling, and immune activation is of special import in cancer. Main Here we review the current understanding of Ca2+ in each of these processes vital to the initiation, spread, and drug resistance of malignancies. We describe the involvement of Ca2+, and Ca2+ related proteins in cell cycle checkpoints and Ca2+ dependent apoptosis and discuss their roles in cellular immortalization. The role of Ca2+ in inter-cellular communication is also discussed in relevance to tumor-stromal communication, angiogenesis, and tumor microinvasion. The role that Ca2+ plays in immune surveillance and evasion is also addressed. Finally, we discuss the possibility of targeting Ca2+ singling to address the most pressing topics of cancer treatment: metastatic disease and drug resistance. Conclusion This review discusses the current understanding of Ca2+ in cancer. By addressing Ca2+ facilitated angiogenesis, immune evasion, metastasis, and drug resistance, we anticipate future avenues for development of Ca2+ as a nexus of therapy.
Collapse
Affiliation(s)
- MengMeng Xu
- 1Medical-Scientist Training Program, Duke University Medical Center, Durham, NC 27710 USA.,2Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Andreas Seas
- 1Medical-Scientist Training Program, Duke University Medical Center, Durham, NC 27710 USA
| | - Musa Kiyani
- 3School of Medicine, Duke University Medical Center, Durham, NC 27710 USA.,4Duke-NUS Medical School, Singapore, 169857 Singapore
| | - Keven S Y Ji
- 3School of Medicine, Duke University Medical Center, Durham, NC 27710 USA
| | - Hannah N Bell
- 1Medical-Scientist Training Program, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
250
|
Abstract
Evidence is increasing on the crucial role of the extracellular matrix (ECM) in breast cancer progression, invasion and metastasis with almost all mortality cases owing to metastasis. The epithelial-mesenchymal transition is the first signal of metastasis involving different transcription factors such as Snail, TWIST, and ZEB1. ECM remodeling is a major event promoting cancer invasion and metastasis; where matrix metalloproteinases (MMPs) such as MMP-2, -9, -11, and -14 play vital roles degrading the matrix proteins for cancer spread. The β-D mannuronic acid (MMP inhibitor) has anti-metastatic properties through inhibition of MMP-2, and -9 and could be a potential therapeutic agent. Besides the MMPs, the enzymes such as LOXL2, LOXL4, procollagen lysyl hydroxylase-2, and heparanase also regulate breast cancer progression. The important ECM proteins like integrins (b1-, b5-, and b6- integrins), ECM1 protein, and Hic-5 protein are also actively involved in breast cancer development. The stromal cells such as tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and adipocytes also contribute in tumor development through different processes. The TAMs become proangiogenic through secretion of VEGF-A and building vessel network for nourishment and invasion of the tumor mass. The latest developments of ECM involvement in breast cancer progression has been discussed in this review and this study will help researchers in designing future work on breast cancer pathogenesis and developing therapy targeted to the ECM components.
Collapse
Affiliation(s)
- Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University , Phagwara, Punjab, 144411, India
| | - Jagadeesh Janjanam
- Department of Developmental Neurobiology , St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| |
Collapse
|