1
|
Wirth F, Zoeller C, Lubosch A, Schroeder-Braunstein J, Wabnitz G, Nakchbandi IA. Insights into the metastatic bone marrow niche gained from fibronectin and β1 integrin transgenic mice. Neoplasia 2024; 58:101058. [PMID: 39413671 PMCID: PMC11530925 DOI: 10.1016/j.neo.2024.101058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 10/18/2024]
Abstract
Tumor cells can migrate from a primary cancer and form metastases by localizing to niches within other organs including the bone marrow, where tumor cells may exploit the hematopoietic stem cell niche. The precise composition of the premetastatic and the hematopoietic niches and the degree of overlap between them remain elusive. Because the extracellular matrix protein fibronectin is expressed in the pre-metastatic lung microenvironment, we evaluated the implications of its loss, as well as those of loss of its primary receptor subunit, β1 integrin, in various bone marrow cell types both in breast cancer bone metastasis and hematopoiesis. Using eight transgenic mouse models, we established that fibronectin production by osterix-expressing marrow cells, or β1 integrin expression (on vav, mx, or leptin receptor expressing cells), affects MDA-MB-231 breast cancer cell numbers in the bone marrow. Additionally, we identified stromal subpopulations that modulate transmigration through blood vessel walls. Not the number of tumor cells, but rather the changes in the microenvironment dictated whether the tumor progresses. Furthermore, hematopoiesis, particularly myelopoiesis, was affected in some of the models showing changes in tumor homing. In conclusion, there is partial overlap between the pre-metastatic and the hematopoietic niches in the bone marrow. Moreover, we have delineated a cascade starting with fibronectin secreted by pre-osteoblastic cells, which potentially acts on β1 integrin in specific stromal cell subsets, thereby inhibiting the formation of new breast cancer lesions in the bone marrow. This work therefore sheds light on the role of various stromal cell subpopulations that influence tumor behavior and affect hematopoiesis.
Collapse
Affiliation(s)
- Franziska Wirth
- Institute of Immunology, Heidelberg University, 69120, Heidelberg, Germany
| | - Caren Zoeller
- Institute of Immunology, Heidelberg University, 69120, Heidelberg, Germany
| | - Alexander Lubosch
- Institute of Immunology, Heidelberg University, 69120, Heidelberg, Germany
| | | | - Guido Wabnitz
- Institute of Immunology, Heidelberg University, 69120, Heidelberg, Germany
| | - Inaam A Nakchbandi
- Institute of Immunology, Heidelberg University, 69120, Heidelberg, Germany; Max-Planck Institute for Biochemistry, 82152, Martinsried, Germany; Max-Planck Institute for Medical Research, 69120, Heidelberg, Germany.
| |
Collapse
|
2
|
Monteran L, Zait Y, Erez N. It's all about the base: stromal cells are central orchestrators of metastasis. Trends Cancer 2024; 10:208-229. [PMID: 38072691 DOI: 10.1016/j.trecan.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 03/16/2024]
Abstract
The tumor microenvironment (TME) is an integral part of tumors and plays a central role in all stages of carcinogenesis and progression. Each organ has a unique and heterogeneous microenvironment, which affects the ability of disseminated cells to grow in the new and sometimes hostile metastatic niche. Resident stromal cells, such as fibroblasts, osteoblasts, and astrocytes, are essential culprits in the modulation of metastatic progression: they transition from being sentinels of tissue integrity to being dysfunctional perpetrators that support metastatic outgrowth. Therefore, better understanding of the complexity of their reciprocal interactions with cancer cells and with other components of the TME is essential to enable the design of novel therapeutic approaches to prevent metastatic relapse.
Collapse
Affiliation(s)
- Lea Monteran
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Zait
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
3
|
Farooq F, Amin A, Wani UM, Lone A, Qadri RA. Shielding and nurturing: Fibronectin as a modulator of cancer drug resistance. J Cell Physiol 2023; 238:1651-1669. [PMID: 37269547 DOI: 10.1002/jcp.31048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/02/2023] [Accepted: 05/10/2023] [Indexed: 06/05/2023]
Abstract
Resistance to chemotherapy and targeted therapies constitute a common hallmark of most cancers and represent a dominant factor fostering tumor relapse and metastasis. Fibronectin, an abundant extracellular matrix glycoprotein, has long been proposed to play an important role in the pathobiology of cancer. Recent research has unraveled the role of Fibronectin in the onset of chemoresistance against a variety of antineoplastic drugs including DNA-damaging agents, hormone receptor antagonists, tyrosine kinase inhibitors, microtubule destabilizing agents, etc. The current review summarizes the role played by Fibronectin in mediating drug resistance against diverse anticancer drugs. We have also discussed how the aberrant expression of Fibronectin drives the oncogenic signaling pathways ultimately leading to drug resistance through the inhibition of apoptosis, promotion of cancer cell growth and proliferation.
Collapse
Affiliation(s)
- Faizah Farooq
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Asif Amin
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Umer Majeed Wani
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Asif Lone
- Department of Biochemistry, Deshbandu College, University of Delhi, Delhi, India
| | - Raies A Qadri
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
4
|
Chen S, He T, Zhong Y, Chen M, Yao Q, Chen D, Shao Z, Xiao G. Roles of focal adhesion proteins in skeleton and diseases. Acta Pharm Sin B 2023; 13:998-1013. [PMID: 36970189 PMCID: PMC10031257 DOI: 10.1016/j.apsb.2022.09.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/04/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
The skeletal system, which contains bones, joints, tendons, ligaments and other elements, plays a wide variety of roles in body shaping, support and movement, protection of internal organs, production of blood cells and regulation of calcium and phosphate metabolism. The prevalence of skeletal diseases and disorders, such as osteoporosis and bone fracture, osteoarthritis, rheumatoid arthritis, and intervertebral disc degeneration, increases with age, causing pain and loss of mobility and creating a huge social and economic burden globally. Focal adhesions (FAs) are macromolecular assemblies that are composed of the extracellular matrix (ECM), integrins, intracellular cytoskeleton and other proteins, including kindlin, talin, vinculin, paxillin, pinch, Src, focal adhesion kinase (FAK) and integrin-linked protein kinase (ILK) and other proteins. FA acts as a mechanical linkage connecting the ECM and cytoskeleton and plays a key role in mediating cell-environment communications and modulates important processes, such as cell attachment, spreading, migration, differentiation and mechanotransduction, in different cells in skeletal system by impacting distinct outside-in and inside-out signaling pathways. This review aims to integrate the up-to-date knowledge of the roles of FA proteins in the health and disease of skeletal system and focuses on the specific molecular mechanisms and underlying therapeutic targets for skeletal diseases.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
5
|
Peng Z, Lv X, Huang S. Recent Progress on the Role of Fibronectin in Tumor Stromal Immunity and Immunotherapy. Curr Top Med Chem 2022; 22:2494-2505. [PMID: 35708087 DOI: 10.2174/1568026622666220615152647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 01/20/2023]
Abstract
As a major component of the stromal microenvironment of various solid tumors, the extracellular matrix (ECM) has attracted increasing attention in cancer-related studies. ECM in the tumor stroma not only provides an external barrier and framework for tumor cell adhesion and movement, but also acts as an active regulator that modulates the tumor microenvironment, including stromal immunity. Fibronectin (Fn), as a core component of the ECM, plays a key role in the assembly and remodeling of the ECM. Hence, understanding the role of Fn in the modulation of tumor stromal immunity is of great importance for cancer immunotherapy. Hence, in-depth studies on the underlying mechanisms of Fn in tumors are urgently needed to clarify the current understanding and issues and to identify new and specific targets for effective diagnosis and treatment purposes. In this review, we summarize the structure and role of Fn, its potent derivatives in tumor stromal immunity, and their biological effects and mechanisms in tumor development. In addition, we discuss the novel applications of Fn in tumor treatment. Therefore, this review can provide prospective insight into Fn immunotherapeutic applications in tumor treatment.
Collapse
Affiliation(s)
- Zheng Peng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xiaolan Lv
- Department of Laboratory Medicine, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Shigao Huang
- Department of Radiation Oncology, The First Affiliated Hospital, Air Force Medical University, Xi an, Shaan Xi, China
| |
Collapse
|
6
|
Malara A, Gruppi C, Massa M, Tira ME, Rosti V, Balduini A, Barosi G. Elevated plasma EDA fibronectin in primary myelofibrosis is determined by high allele burden of JAK2V617F mutation and strongly predicts splenomegaly progression. Front Oncol 2022; 12:987643. [PMID: 36212480 PMCID: PMC9532599 DOI: 10.3389/fonc.2022.987643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
In primary myelofibrosis, extra-domain A fibronectin (EDA-FN), the result of alternative splicing of FN gene, sustains megakaryocyte proliferation and confers a pro-inflammatory phenotype to bone marrow cell niches. In this work we assessed the levels of circulating EDA-FN in plasma samples of 122 patients with primary myelofibrosis. Patients with a homozygous JAK2V617F genotype displayed the higher level of plasma EDA-FN. Increased EDA-FN levels were associated with anemia, elevated high-sensitivity C-reactive protein, bone marrow fibrosis and splanchnic vein thrombosis at diagnosis. While no correlation was observed with CD34+ hematopoietic stem cell mobilization, elevated blood level of EDA-FN at diagnosis was a predictor of large splenomegaly (over 10 cm from the left costal margin) outcome. Thus, EDA-FN expression in primary myelofibrosis may represent the first marker of disease progression, and a novel target to treat splenomegaly.
Collapse
Affiliation(s)
- Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Alessandro Malara, ; Alessandra Balduini,
| | - Cristian Gruppi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Margherita Massa
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Maria Enrica Tira
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Alessandro Malara, ; Alessandra Balduini,
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| |
Collapse
|
7
|
Martín-Otal C, Lasarte-Cia A, Serrano D, Casares N, Conde E, Navarro F, Sánchez-Moreno I, Gorraiz M, Sarrión P, Calvo A, De Andrea CE, Echeveste J, Vilas A, Rodriguez-Madoz JR, San Miguel J, Prosper F, Hervas-Stubbs S, Lasarte JJ, Lozano T. Targeting the extra domain A of fibronectin for cancer therapy with CAR-T cells. J Immunother Cancer 2022; 10:jitc-2021-004479. [PMID: 35918123 PMCID: PMC9351345 DOI: 10.1136/jitc-2021-004479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND One of the main difficulties of adoptive cell therapies with chimeric antigen receptor (CAR)-T cells in solid tumors is the identification of specific target antigens. The tumor microenvironment can present suitable antigens for CAR design, even though they are not expressed by the tumor cells. We have generated a CAR specific for the splice variant extra domain A (EDA) of fibronectin, which is highly expressed in the tumor stroma of many types of tumors but not in healthy tissues. METHODS EDA expression was explored in RNA-seq data from different human tumor types and by immunohistochemistry in paraffin-embedded tumor biopsies. Murine and human anti-EDA CAR-T cells were prepared using recombinant retro/lentiviruses, respectively. The functionality of EDA CAR-T cells was measured in vitro in response to antigen stimulation. The antitumor activity of EDA CAR-T cells was measured in vivo in C57BL/6 mice challenged with PM299L-EDA hepatocarcinoma cell line, in 129Sv mice-bearing F9 teratocarcinoma and in NSG mice injected with the human hepatocarcinoma cell line PLC. RESULTS EDA CAR-T cells recognized and killed EDA-expressing tumor cell lines in vitro and rejected EDA-expressing tumors in immunocompetent mice. Notably, EDA CAR-T cells showed an antitumor effect in mice injected with EDA-negative tumor cells lines when the tumor stroma or the basement membrane of tumor endothelial cells express EDA. Thus, EDA CAR-T administration delayed tumor growth in immunocompetent 129Sv mice challenged with teratocarcinoma cell line F9. EDA CAR-T treatment exerted an antiangiogenic effect and significantly reduced gene signatures associated with epithelial-mesenchymal transition, collagen synthesis, extracellular matrix organization as well as IL-6-STAT5 and KRAS pathways. Importantly, the human version of EDA CAR, that includes the human 41BB and CD3ζ endodomains, exerted strong antitumor activity in NSG mice challenged with the human hepatocarcinoma cell line PLC, which expresses EDA in the tumor stroma and the endothelial vasculature. EDA CAR-T cells exhibited a tropism for EDA-expressing tumor tissue and no toxicity was observed in tumor bearing or in healthy mice. CONCLUSIONS These results suggest that targeting the tumor-specific fibronectin splice variant EDA with CAR-T cells is feasible and offers a therapeutic option that is applicable to different types of cancer.
Collapse
Affiliation(s)
- Celia Martín-Otal
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Aritz Lasarte-Cia
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Diego Serrano
- Programa de Tumores sólidos, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Noelia Casares
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Enrique Conde
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Flor Navarro
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Inés Sánchez-Moreno
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Marta Gorraiz
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Patricia Sarrión
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Alfonso Calvo
- Programa de Tumores sólidos, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Carlos E De Andrea
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.,Departamento de Patología, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - José Echeveste
- Departamento de Patología, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Amaia Vilas
- Programa de Hemato-Oncología, Centro de Investigación Médica Aplicada, CIMA, Pamplona, Spain
| | - Juan Roberto Rodriguez-Madoz
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain,Cancer Center Universidad de Navarra (CCUN), Universidad de Navarra, Pamplona, Spain
| | - Jesús San Miguel
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain,Cancer Center Universidad de Navarra (CCUN), Universidad de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.,Programa de Hemato-Oncología, Centro de Investigación Médica Aplicada, CIMA, Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain,Cancer Center Universidad de Navarra (CCUN), Universidad de Navarra, Pamplona, Spain
| | - Sandra Hervas-Stubbs
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Juan Jose Lasarte
- Departamento de Hematología, Clínica Universidad de Navarra, Pamplona, Spain,Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Teresa Lozano
- Departamento de Hematología, Clínica Universidad de Navarra, Pamplona, Spain,Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
8
|
Soubeyrand S, Lau P, Nikpay M, Dang AT, McPherson R. Common Polymorphism That Protects From Cardiovascular Disease Increases Fibronectin Processing and Secretion. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2022; 15:e003428. [PMID: 35130031 DOI: 10.1161/circgen.121.003428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Fibronectin (FN1) is an essential regulator of homodynamic processes and tissue remodeling that have been proposed to contribute to atherosclerosis. Moreover, recent large-scale genome-wide association studies (GWAS) have linked common genetic variants within the FN1 gene to coronary artery disease risk. METHODS Public databases were analyzed by 2-Sample Mendelian Randomization. Expression constructs encoding short FN1 reporter constructs and full-length plasma FN1 variants were introduced in various cell models. Secreted and cellular levels were then analyzed and quantified by SDS-PAGE and fluorescence microscopy. Mass spectrometry and glycosylation analyses were performed to probe possible posttranscriptional differences. RESULTS Bioinformatic analyses revealed that common coronary artery disease risk single nucleotide polymorphisms in the FN1 locus associate with circulating levels of FN1 and that higher FN1 (fibronectin 1) protein levels in plasma are linked to lower coronary artery disease risk. The coronary artery disease-associated FN1 locus encompasses a common polymorphism that translates a L15Q variant situated within the FN1 signal peptide. Introduction of FN1 reporter constructs, differing at position 15, revealed differences in secretion, with the FN1 Q15 variant being less well secreted. Moreover, the L15Q polymorphism was found to alter glycosylation in some cell models but not in human plasma. CONCLUSIONS In addition to providing novel functional evidence implicating FN1 in cardioprotection, these findings demonstrate that a common variant within a secretion signal peptide regulates protein function.
Collapse
Affiliation(s)
- Sébastien Soubeyrand
- Atherogenomics Laboratory (S.S., P.L., M.N., A.-T.D., R.M.), University of Ottawa Heart Institute, Canada
| | - Paulina Lau
- Atherogenomics Laboratory (S.S., P.L., M.N., A.-T.D., R.M.), University of Ottawa Heart Institute, Canada
| | - Majid Nikpay
- Atherogenomics Laboratory (S.S., P.L., M.N., A.-T.D., R.M.), University of Ottawa Heart Institute, Canada
| | - Anh-Thu Dang
- Atherogenomics Laboratory (S.S., P.L., M.N., A.-T.D., R.M.), University of Ottawa Heart Institute, Canada
| | - Ruth McPherson
- Atherogenomics Laboratory (S.S., P.L., M.N., A.-T.D., R.M.), University of Ottawa Heart Institute, Canada.,Department of Medicine, Ruddy Canadian Cardiovascular Genetics Centre (R.M.), University of Ottawa Heart Institute, Canada
| |
Collapse
|
9
|
Mancini SJC, Balabanian K, Corre I, Gavard J, Lazennec G, Le Bousse-Kerdilès MC, Louache F, Maguer-Satta V, Mazure NM, Mechta-Grigoriou F, Peyron JF, Trichet V, Herault O. Deciphering Tumor Niches: Lessons From Solid and Hematological Malignancies. Front Immunol 2021; 12:766275. [PMID: 34858421 PMCID: PMC8631445 DOI: 10.3389/fimmu.2021.766275] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Knowledge about the hematopoietic niche has evolved considerably in recent years, in particular through in vitro analyzes, mouse models and the use of xenografts. Its complexity in the human bone marrow, in particular in a context of hematological malignancy, is more difficult to decipher by these strategies and could benefit from the knowledge acquired on the niches of solid tumors. Indeed, some common features can be suspected, since the bone marrow is a frequent site of solid tumor metastases. Recent research on solid tumors has provided very interesting information on the interactions between tumoral cells and their microenvironment, composed notably of mesenchymal, endothelial and immune cells. This review thus focuses on recent discoveries on tumor niches that could help in understanding hematopoietic niches, with special attention to 4 particular points: i) the heterogeneity of carcinoma/cancer-associated fibroblasts (CAFs) and mesenchymal stem/stromal cells (MSCs), ii) niche cytokines and chemokines, iii) the energy/oxidative metabolism and communication, especially mitochondrial transfer, and iv) the vascular niche through angiogenesis and endothelial plasticity. This review highlights actors and/or pathways of the microenvironment broadly involved in cancer processes. This opens avenues for innovative therapeutic opportunities targeting not only cancer stem cells but also their regulatory tumor niche(s), in order to improve current antitumor therapies.
Collapse
Affiliation(s)
- Stéphane J C Mancini
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMR1236, Rennes 1 University, Etablissement Français du Sang Bretagne, Rennes, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France
| | - Karl Balabanian
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Saint-Louis Research Institute, University of Paris, EMiLy, INSERM U1160, Paris, France.,The Organization for Partnerships in Leukemia (OPALE) Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France
| | - Isabelle Corre
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), Signaling in Oncogenesis Angiogenesis and Permeability (SOAP), INSERM UMR1232, Centre National de la Recherche scientifique (CNRS) ERL600, Université de Nantes, Nantes, France
| | - Julie Gavard
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), Signaling in Oncogenesis Angiogenesis and Permeability (SOAP), INSERM UMR1232, Centre National de la Recherche scientifique (CNRS) ERL600, Université de Nantes, Nantes, France.,Integrated Center for Oncology, St. Herblain, France
| | - Gwendal Lazennec
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Centre National de la Recherche scientifique (CNRS) UMR9005, SYS2DIAG-ALCEDIAG, Montpellier, France
| | - Marie-Caroline Le Bousse-Kerdilès
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMRS-MD1197, Paris-Saclay University, Paul-Brousse Hospital, Villejuif, France
| | - Fawzia Louache
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMRS-MD1197, Paris-Saclay University, Paul-Brousse Hospital, Villejuif, France
| | - Véronique Maguer-Satta
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancer Research Center of Lyon (CRCL), CNRS UMR5286, INSERM U1052, Lyon 1 university, Lean Bérard Center, Lyon, France
| | - Nathalie M Mazure
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM U1065, C3M, University of Côte d'Azur (UCA), Nice, France
| | - Fatima Mechta-Grigoriou
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Stress and Cancer Laboratory, Institut Curie, INSERM U830, Paris Sciences et Lettres (PSL) Research University, Team Babelized Ligue Nationale Contre le Cancer (LNCC), Paris, France
| | - Jean-François Peyron
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM U1065, C3M, University of Côte d'Azur (UCA), Nice, France
| | - Valérie Trichet
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,INSERM UMR1238 Phy-Os, Université de Nantes, Nantes, France
| | - Olivier Herault
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,The Organization for Partnerships in Leukemia (OPALE) Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France.,Centre National de la Recherche scientifique (CNRS) ERL7001 LNOx, EA7501, Tours University, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| |
Collapse
|
10
|
Ling Z, Yang C, Tan J, Dou C, Chen Y. Beyond immunosuppressive effects: dual roles of myeloid-derived suppressor cells in bone-related diseases. Cell Mol Life Sci 2021; 78:7161-7183. [PMID: 34635950 PMCID: PMC11072300 DOI: 10.1007/s00018-021-03966-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/11/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells (IMCs) with immunosuppressive functions, whereas IMCs originally differentiate into granulocytes, macrophages, and dendritic cells (DCs) to participate in innate immunity under steady-state conditions. At present, difficulties remain in identifying MDSCs due to lacking of specific biomarkers. To make identification of MDSCs accurately, it also needs to be determined whether having immunosuppressive functions. MDSCs play crucial roles in anti-tumor, angiogenesis, and metastasis. Meanwhile, MDSCs could make close interaction with osteoclasts, osteoblasts, chondrocytes, and other stromal cells within microenvironment of bone and joint, and thereby contributing to poor prognosis of bone-related diseases such as cancer-related bone metastasis, osteosarcoma (OS), rheumatoid arthritis (RA), osteoarthritis (OA), and orthopedic trauma. In addition, MDSCs have been shown to participate in the procedure of bone repair. In this review, we have summarized the function of MDSCs in cancer-related bone metastasis, the interaction with stromal cells within the bone microenvironment as well as joint microenvironment, and the critical role of MDSCs in bone repair. Besides, the promising value of MDSCs in the treatment for bone-related diseases is also well discussed.
Collapse
Affiliation(s)
- Zhiguo Ling
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chuan Yang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiulin Tan
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ce Dou
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yueqi Chen
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
11
|
Wirth F, Huck K, Lubosch A, Zoeller C, Ghura H, Porubsky S, Nakchbandi IA. Cdc42 in osterix-expressing cells alters osteoblast behavior and myeloid lineage commitment. Bone 2021; 153:116150. [PMID: 34400384 DOI: 10.1016/j.bone.2021.116150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023]
Abstract
Osteoblasts are not only responsible for bone formation. They also support hematopoiesis. This requires responding to cues originating from several signaling pathways, a task performed by Rho GTPases. We therefore examined several transgenic mouse models and used inhibitors of Cdc42 in vitro. Deletion of Cdc42 in vivo using the Osterix promoter suppressed osteoblast function, while its deletion in differentiating osteoblasts using the Collagen-α1(I) promoter decreased osteoblast numbers. In both cases, bone mineral density diminished confirming the importance of Cdc42. Evaluation of hematopoiesis revealed that deletion of Cdc42 using the Osterix, but not the Collagen-α1(I) promoter increased the common myeloid progenitors (CMPs) in the bone marrow as well as the erythrocytes and the thrombocytes/platelets in peripheral blood. Causality between Cdc42 loss in early osteoblasts and increased myelopoiesis was confirmed in vitro. Work in vitro supported the conclusion that interleukin-4 mediated the increase in myelopoiesis. Thus, Cdc42 is required for healthy bone through regulation of bone formation in Osterix-expressing osteoblasts and the number of osteoblasts in differentiating osteoblasts. In addition, its expression in early osteoblasts/stromal cells modulates myelopoiesis. This highlights the importance of osteoblasts in regulating hematopoiesis.
Collapse
Affiliation(s)
- Franziska Wirth
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Katrin Huck
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Alexander Lubosch
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Caren Zoeller
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Hiba Ghura
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Stefan Porubsky
- Institute of Pathology, University of Mainz, 55131 Mainz, Germany
| | - Inaam A Nakchbandi
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany; Max-Planck Institute for Biochemistry, 82152 Martinsried, Germany.
| |
Collapse
|
12
|
Hofbauer LC, Bozec A, Rauner M, Jakob F, Perner S, Pantel K. Novel approaches to target the microenvironment of bone metastasis. Nat Rev Clin Oncol 2021; 18:488-505. [PMID: 33875860 DOI: 10.1038/s41571-021-00499-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Bone metastases are a frequent and severe complication of advanced-stage cancers. Breast and prostate cancers, the most common malignancies in women and men, respectively, have a particularly high propensity to metastasize to bone. Conceptually, circulating tumour cells (CTCs) in the bloodstream and disseminated tumour cells (DTCs) in the bone marrow provide a snapshot of the dissemination and colonization process en route to clinically apparent bone metastases. Many cell types that constitute the bone microenvironment, including osteoblasts, osteocytes, osteoclasts, adipocytes, endothelial cells, haematopoietic stem cells and immune cells, engage in a dialogue with tumour cells. Some of these cells modify tumour biology, while others are disrupted and out-competed by tumour cells, thus leading to distinct phases of tumour cell migration, dormancy and latency, and therapy resistance and progression to overt bone metastases. Several current bone-protective therapies act by interrupting these interactions, mainly by targeting tumour cell-osteoclast interactions. In this Review, we describe the functional roles of the bone microenvironment and its components in the initiation and propagation of skeletal metastases, outline the biology and clinical relevance of CTCs and DTCs, and discuss established and future therapeutic approaches that specifically target defined components of the bone microenvironment to prevent or treat skeletal metastases.
Collapse
Affiliation(s)
- Lorenz C Hofbauer
- University Center for Healthy Aging, Dresden University of Technology, Dresden, Germany. .,Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) partner site Dresden, Dresden, Germany.
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Martina Rauner
- University Center for Healthy Aging, Dresden University of Technology, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Franz Jakob
- Department of Orthopedic Surgery, Julius Maximilians University of Würzburg, Würzburg, Germany.,Department of Functional Materials in Medicine and Dentistry, Julius Maximilians University of Würzburg, Würzburg, Germany
| | - Sven Perner
- Institute of Pathology, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.,Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
13
|
Myofibroblasts: Function, Formation, and Scope of Molecular Therapies for Skin Fibrosis. Biomolecules 2021; 11:biom11081095. [PMID: 34439762 PMCID: PMC8391320 DOI: 10.3390/biom11081095] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022] Open
Abstract
Myofibroblasts are contractile, α-smooth muscle actin-positive cells with multiple roles in pathophysiological processes. Myofibroblasts mediate wound contractions, but their persistent presence in tissues is central to driving fibrosis, making them attractive cell targets for the development of therapeutic treatments. However, due to shared cellular markers with several other phenotypes, the specific targeting of myofibroblasts has long presented a scientific and clinical challenge. In recent years, myofibroblasts have drawn much attention among scientific research communities from multiple disciplines and specialisations. As further research uncovers the characterisations of myofibroblast formation, function, and regulation, the realisation of novel interventional routes for myofibroblasts within pathologies has emerged. The research community is approaching the means to finally target these cells, to prevent fibrosis, accelerate scarless wound healing, and attenuate associated disease-processes in clinical settings. This comprehensive review article describes the myofibroblast cell phenotype, their origins, and their diverse physiological and pathological functionality. Special attention has been given to mechanisms and molecular pathways governing myofibroblast differentiation, and updates in molecular interventions.
Collapse
|
14
|
Ghura H, Keimer M, von Au A, Hackl N, Klemis V, Nakchbandi IA. Inhibition of fibronectin accumulation suppresses tumor growth. Neoplasia 2021; 23:837-850. [PMID: 34298233 PMCID: PMC8322122 DOI: 10.1016/j.neo.2021.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Understanding how the extracellular matrix affects cancer development constitutes an emerging research field. Fibronectin and collagen are two intriguing matrix components found in cancer. Large concentrations of fibronectin or collagen type I have been implicated in poor prognosis in patients. In a mouse model, we had shown that genetically decreasing circulating fibronectin resulted in smaller tumors. We therefore aimed to manipulate fibronectin pharmacologically and determine how cancer development is affected. Deletion of fibronectin in human breast cancer cells (MDA-MB-231) using shRNA (knockdown: Kd) improved survival and diminished tumor burden in a model of metastatic lesions and in a model of local growth. Based on these findings, it seemed reasonable to attempt to prevent fibronectin accumulation using a bacterial derived peptide called pUR4. Treatment with this peptide for 10 days in the breast cancer local growth model or for 5 days in a melanoma skin cancer model (B16) was associated with a significant suppression of cancer growth. Treatment aimed at inhibiting collagen type I accumulation without interfering with fibronectin could not affect any changes in vivo. In the absence of fibronectin, diminished cancer progression was due to inhibition of proliferation, even though changes in blood vessels were also detected. Decreased proliferation could be attributed to decreased ERK phosphorylation and diminished YAP expression. In summary, manipulating fibronectin diminishes cancer progression, mostly by suppressing cell proliferation. This suggests that matrix modulation could be used as an adjuvant to conventional therapy as long as a decrease in fibronectin is obtained.
Collapse
Affiliation(s)
- Hiba Ghura
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Marin Keimer
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Anja von Au
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Norman Hackl
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Verena Klemis
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Inaam A Nakchbandi
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany; Max-Planck Institute for Medical Research, Heidelberg, Germany; Max-Planck Institute for Biochemistry, Martinsried, Germany.
| |
Collapse
|
15
|
Fibronectin in development and wound healing. Adv Drug Deliv Rev 2021; 170:353-368. [PMID: 32961203 DOI: 10.1016/j.addr.2020.09.005] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/26/2020] [Accepted: 09/15/2020] [Indexed: 01/15/2023]
Abstract
Fibronectin structure and composition regulate contextual cell signaling. Recent advances have been made in understanding fibronectin and its role in tissue organization and repair. This review outlines fibronectin splice variants and their functions, evaluates potential therapeutic strategies targeting or utilizing fibronectin, and concludes by discussing potential future directions to modulate fibronectin function in development and wound healing.
Collapse
|
16
|
Zhang L, Yan H, Tai Y, Xue Y, Wei Y, Wang K, Zhao Q, Wang S, Kong D, Midgley AC. Design and Evaluation of a Polypeptide that Mimics the Integrin Binding Site for EDA Fibronectin to Block Profibrotic Cell Activity. Int J Mol Sci 2021; 22:ijms22041575. [PMID: 33557232 PMCID: PMC7913925 DOI: 10.3390/ijms22041575] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is characterized by excessive production of disorganized collagen- and fibronectin-rich extracellular matrices (ECMs) and is driven by the persistence of myofibroblasts within tissues. A key protein contributing to myofibroblast differentiation is extra domain A fibronectin (EDA-FN). We sought to target and interfere with interactions between EDA-FN and its integrin receptors to effectively inhibit profibrotic activity and myofibroblast formation. Molecular docking was used to assist in the design of a blocking polypeptide (antifibrotic 38-amino-acid polypeptide, AF38Pep) for specific inhibition of EDA-FN associations with the fibroblast-expressed integrins α4β1 and α4β7. Blocking peptides were designed and evaluated in silico before synthesis, confirmation of binding specificity, and evaluation in vitro. We identified the high-affinity EDA-FN C-C′ loop binding cleft within integrins α4β1 and α4β7. The polypeptide with the highest predicted binding affinity, AF38Pep, was synthesized and could achieve specific binding to myofibroblast fibronectin-rich ECM and EDA-FN C-C′ loop peptides. AF38Pep demonstrated potent myofibroblast inhibitory activity at 10 µg/mL and was not cytotoxic. Treatment with AF38Pep prevented integrin α4β1-mediated focal adhesion kinase (FAK) activation and early signaling through extracellular-signal-regulated kinases 1 and 2 (ERK1/2), attenuated the expression of pro-matrix metalloproteinase 9 (MMP9) and pro-MMP2, and inhibited collagen synthesis and deposition. Immunocytochemistry staining revealed an inhibition of α-smooth muscle actin (α-SMA) incorporation into actin stress fibers and attenuated cell contraction. Increases in the expression of mRNA associated with fibrosis and downstream from integrin signaling were inhibited by treatment with AF38Pep. Our study suggested that AF38Pep could successfully interfere with EDA-FN C-C′ loop-specific integrin interactions and could act as an effective inhibitor of fibroblast of myofibroblast differentiation.
Collapse
Affiliation(s)
- Lin Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
| | - Hongyu Yan
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
| | - Yifan Tai
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
| | - Yueming Xue
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
| | - Yongzhen Wei
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
| | - Kai Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
| | - Qiang Zhao
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
| | - Shufang Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
- Correspondence: (S.W.); (A.C.M.); Tel.: +86-1562-004-7851 (A.C.M.)
| | - Deling Kong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
- Rongxiang Xu Center for Regenerative Life Science, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Adam C. Midgley
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (L.Z.); (H.Y.); (Y.T.); (Y.X.); (Y.W.); (K.W.); (Q.Z.); (D.K.)
- Rongxiang Xu Center for Regenerative Life Science, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
- Correspondence: (S.W.); (A.C.M.); Tel.: +86-1562-004-7851 (A.C.M.)
| |
Collapse
|
17
|
Stromal Protein-Mediated Immune Regulation in Digestive Cancers. Cancers (Basel) 2021; 13:cancers13010146. [PMID: 33466303 PMCID: PMC7795083 DOI: 10.3390/cancers13010146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Solid cancers are surrounded by a network of non-cancerous cells comprising different cell types, including fibroblasts, and acellular protein structures. This entire network is called the tumor microenvironment (TME) and it provides a physical barrier to the tumor shielding it from infiltrating immune cells, such as lymphocytes, or therapeutic agents. In addition, the TME has been shown to dampen efficient immune responses of infiltrated immune cells, which are key in eliminating cancer cells from the organism. In this review, we will discuss how TME proteins in particular are involved in this dampening effect, known as immunosuppression. We will focus on three different types of digestive cancers: pancreatic cancer, colorectal cancer, and gastric cancer. Moreover, we will discuss current therapeutic approaches using TME proteins as targets to reverse their immunosuppressive effects. Abstract The stromal tumor microenvironment (TME) consists of immune cells, vascular and neural structures, cancer-associated fibroblasts (CAFs), as well as extracellular matrix (ECM), and favors immune escape mechanisms promoting the initiation and progression of digestive cancers. Numerous ECM proteins released by stromal and tumor cells are crucial in providing physical rigidity to the TME, though they are also key regulators of the immune response against cancer cells by interacting directly with immune cells or engaging with immune regulatory molecules. Here, we discuss current knowledge of stromal proteins in digestive cancers including pancreatic cancer, colorectal cancer, and gastric cancer, focusing on their functions in inhibiting tumor immunity and enabling drug resistance. Moreover, we will discuss the implication of stromal proteins as therapeutic targets to unleash efficient immunotherapy-based treatments.
Collapse
|
18
|
Amin A, Mokhdomi TA, Bukhari S, Wani Z, Chikan NA, Shah BA, Koul AM, Majeed U, Farooq F, Qadri A, Qadri RA. Lung cancer cell-derived EDA-containing fibronectin induces an inflammatory response from monocytes and promotes metastatic tumor microenvironment. J Cell Biochem 2021; 122:562-576. [PMID: 33393138 DOI: 10.1002/jcb.29883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/25/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022]
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in facilitating tumor growth and metastasis. This tumor-promoting propensity of TAMs sets in as a result of their complex cross-talk with tumor cells mediated primarily by tumor cell-secreted proteins in the tumor microenvironment. To explore such interactions, we employed an immunoscreening approach involving the immunization of Balb-c mice with model human lung carcinoma cell line, A549. From serological examination combined with mass spectrometric analysis, EDA-containing fibronectin (EDAFN ) was identified as a conspicuous immunogenic protein in A549 cell secretome. We showed that A549 secreted EDAFN engages TLR-4 on THP-1 monocytes to drive the proinflammatory response via NF-κB signaling cascade. Conversely, A549 derived EDAFN potentiates their metastatic capacity by inducing epithelial-mesenchymal transition through its autocrine activity. In conclusion, the study proposes a possible mechanism of cellular cross-talk between lung cancer cells and associated monocytes mediated by lung cancer-derived EDAFN and resulting in the establishment of proinflammatory and metastatic tumor microenvironment.
Collapse
Affiliation(s)
- Asif Amin
- Department of Biotechnology, University of Kashmir, Srinagar, India.,Hybridoma Lab, National Institute of Immunology, New Delhi, India
| | - Taseem A Mokhdomi
- Department of Biotechnology, University of Kashmir, Srinagar, India.,Daskdān Innovations Pvt. Ltd., Srinagar, India
| | - Shoiab Bukhari
- Department of Biotechnology, University of Kashmir, Srinagar, India.,The Columbia Center for Translational Immunology, Columbia University Medical Center, New York, USA
| | - Zubair Wani
- Department of Biotechnology, University of Kashmir, Srinagar, India
| | | | - Basit A Shah
- Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Aabid M Koul
- Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Umer Majeed
- Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Faizah Farooq
- Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Ayub Qadri
- Hybridoma Lab, National Institute of Immunology, New Delhi, India
| | - Raies A Qadri
- Department of Biotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
19
|
Wirth F, Lubosch A, Hamelmann S, Nakchbandi IA. Fibronectin and Its Receptors in Hematopoiesis. Cells 2020; 9:cells9122717. [PMID: 33353083 PMCID: PMC7765895 DOI: 10.3390/cells9122717] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/08/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Fibronectin is a ubiquitous extracellular matrix protein that is produced by many cell types in the bone marrow and distributed throughout it. Cells of the stem cell niche produce the various isoforms of this protein. Fibronectin not only provides the cells a scaffold to bind to, but it also modulates their behavior by binding to receptors on the adjacent hematopoietic stem cells and stromal cells. These receptors, which include integrins such as α4β1, α9β1, α4β7, α5β1, αvβ3, Toll-like receptor-4 (TLR-4), and CD44, are found on the hematopoietic stem cell. Because the knockout of fibronectin is lethal during embryonal development and because fibronectin is produced by almost all cell types in mammals, the study of its role in hematopoiesis is difficult. Nevertheless, strong and direct evidence exists for its stimulation of myelopoiesis and thrombopoiesis using in vivo models. Other reviewed effects can be deduced from the study of fibronectin receptors, which showed their activation modifies the behavior of hematopoietic stem cells. Erythropoiesis was only stimulated under hemolytic stress, and mostly late stages of lymphocytic differentiation were modulated. Because fibronectin is ubiquitously expressed, these interactions in health and disease need to be taken into account whenever any molecule is evaluated in hematopoiesis.
Collapse
Affiliation(s)
- Franziska Wirth
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; (F.W.); (A.L.); (S.H.)
| | - Alexander Lubosch
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; (F.W.); (A.L.); (S.H.)
| | - Stefan Hamelmann
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; (F.W.); (A.L.); (S.H.)
| | - Inaam A. Nakchbandi
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; (F.W.); (A.L.); (S.H.)
- Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-622-156-8744
| |
Collapse
|
20
|
Greenwell JC, Torres-Gonzalez E, Ritzenthaler JD, Roman J. Interplay between aging, lung inflammation/remodeling, and fibronectin EDA in lung cancer progression. Cancer Biol Ther 2020; 21:1109-1118. [PMID: 33222614 DOI: 10.1080/15384047.2020.1831372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Lung cancer remains the leading cause of cancer death in the United States. Since most lung cancers occur in aged individuals with chronic lung disorders characterized by inflammation and/or fibrosis, we hypothesized that aging and tissue inflammation/remodeling act in concert to promote lung cancer progression. To test this, we engaged in studies using young and aged C57BL/6 mice in conjunction with bleomycin treatment in a syngeneic model of lung cancer. Wildtype young (3 months) and aged (9 months) C57BL/6 mice were injected with Lewis Lung Carcinoma (LLC) cells at day 14 after injection with phosphate-buffered saline or bleomycin. Untreated aged mice were found to develop more lung metastases than young mice. Bleomycin induced weight loss and lung inflammation/remodeling in both young and aged mice, and it increased the number of lung metastases in aged lungs, but not in young lungs. Since aged lungs show alterations in the expression of fibronectin EDA, we repeated studies in aged WT and aged FN EDA KO mice. In the absence of tissue remodeling/inflammation, WT and FN EDA KO mice developed the same number of metastases when injected with LLC cells. However, the increase in lung metastasis due to bleomycin treatment was abolished in FN EDA KO mice, but only in aged and injured lungs. Together, these studies show increased lung cancer metastasis in aging animals and point to the influence of FN EDA and injury in this process.
Collapse
Affiliation(s)
- John C Greenwell
- Department of Pharmacology & Toxicology, University of Louisville, Health Sciences Center , Louisville, KY, USA
| | - Edilson Torres-Gonzalez
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Disorders, University of Louisville Health Sciences Center , Louisville, KY, USA.,Department of Medicine, Division of Pulmonary, Allergy, and Critical Care and the Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University , Philadelphia, PA, USA
| | - Jeffrey D Ritzenthaler
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Disorders, University of Louisville Health Sciences Center , Louisville, KY, USA.,Department of Medicine, Division of Pulmonary, Allergy, and Critical Care and the Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University , Philadelphia, PA, USA
| | - Jesse Roman
- Department of Pharmacology & Toxicology, University of Louisville, Health Sciences Center , Louisville, KY, USA.,Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Disorders, University of Louisville Health Sciences Center , Louisville, KY, USA.,Department of Medicine, Division of Pulmonary, Allergy, and Critical Care and the Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University , Philadelphia, PA, USA.,Robley Rex VA Medical Center , Louisville, KY, USA
| |
Collapse
|
21
|
Abstract
Fibronectin (FN) is a large glycoprotein that plays a diverse set of biological roles. This chapter discusses the structural biology, the normal biological functions, and the molecular role of FN and its splice variants in cancer cell proliferation, metastasis, and chemoresistance. The potential role of FN in cancer imaging is discussed in detail. The chapter also discusses the future directions of basic and translational research of fibronectin in the context of the tumor microenvironment and its role in tumor biology.
Collapse
|
22
|
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: 141] [Impact Index Per Article: 23.5] [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
|
23
|
Devignes CS, Aslan Y, Provot S. The systemic control of cancers by the osteoblasts. Oncoscience 2018; 5:167-168. [PMID: 30035179 PMCID: PMC6049310 DOI: 10.18632/oncoscience.437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 04/30/2018] [Indexed: 12/03/2022] Open
Affiliation(s)
| | - Yetki Aslan
- INSERM, Hôpital Lariboisière - Centre Viggo Petersen, 75010 Paris, France
| | - Sylvain Provot
- INSERM, Hôpital Lariboisière - Centre Viggo Petersen, 75010 Paris, France
| |
Collapse
|
24
|
HIF signaling in osteoblast-lineage cells promotes systemic breast cancer growth and metastasis in mice. Proc Natl Acad Sci U S A 2018; 115:E992-E1001. [PMID: 29339479 DOI: 10.1073/pnas.1718009115] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Bone metastasis involves dynamic interplay between tumor cells and the local stromal environment. In bones, local hypoxia and activation of the hypoxia-inducible factor (HIF)-1α in osteoblasts are essential to maintain skeletal homeostasis. However, the role of osteoblast-specific HIF signaling in cancer metastasis is unknown. Here, we show that osteoprogenitor cells (OPCs) are located in hypoxic niches in the bone marrow and that activation of HIF signaling in these cells increases bone mass and favors breast cancer metastasis to bone locally. Remarkably, HIF signaling in osteoblast-lineage cells also promotes breast cancer growth and dissemination remotely, in the lungs and in other tissues distant from bones. Mechanistically, we found that activation of HIF signaling in OPCs increases blood levels of the chemokine C-X-C motif ligand 12 (CXCL12), which leads to a systemic increase of breast cancer cell proliferation and dissemination through direct activation of the CXCR4 receptor. Hence, our data reveal a previously unrecognized role of the hypoxic osteogenic niche in promoting tumorigenesis beyond the local bone microenvironment. They also support the concept that the skeleton is an important regulator of the systemic tumor environment.
Collapse
|
25
|
Rossnagl S, Ghura H, Groth C, Altrock E, Jakob F, Schott S, Wimberger P, Link T, Kuhlmann JD, Stenzl A, Hennenlotter J, Todenhöfer T, Rojewski M, Bieback K, Nakchbandi IA. A Subpopulation of Stromal Cells Controls Cancer Cell Homing to the Bone Marrow. Cancer Res 2017; 78:129-142. [PMID: 29066511 DOI: 10.1158/0008-5472.can-16-3507] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/26/2017] [Accepted: 10/18/2017] [Indexed: 11/16/2022]
Abstract
Breast and prostate cancer cells home to the bone marrow, where they presumably hijack the hematopoietic stem cell niche. We characterize here the elusive premetastatic niche by examining the role of mesenchymal stromal cells (MSC) in cancer cell homing. Decreasing the number of MSC pharmacologically enhanced cancer cell homing to the bone marrow in mice. In contrast, increasing the number of these MSCs by various interventions including G-CSF administration diminished cancer cell homing. The MSC subpopulation that correlated best with cancer cells expressed stem, endothelial, and pericytic cell markers, suggesting these cells represent an undifferentiated component of the niche with vascular commitment. In humans, a MSC subpopulation carrying markers for endothelial and pericytic cells was lower in the presence of cytokeratin+ cells in bone marrow. Taken together, our data show that a subpopulation of MSC with both endothelial and pericytic cell surface markers suppresses the homing of cancer cells to the bone marrow. Similar to the presence of cytokeratin+ cells in the bone marrow, this MSC subpopulation could prove useful in determining the risk of metastatic disease, and its manipulation might offer a new possibility for diminishing bone metastasis formation.Significance: These findings establish an inverse relationship between a subpopulation of mesenchymal stromal cells and cancer cells in the bone marrow. Cancer Res; 78(1); 129-42. ©2017 AACR.
Collapse
Affiliation(s)
- Stephanie Rossnagl
- Max-Planck Institute for Biochemistry, Martinsried, Germany.,Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Hiba Ghura
- Max-Planck Institute for Biochemistry, Martinsried, Germany.,Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Christopher Groth
- Max-Planck Institute for Biochemistry, Martinsried, Germany.,Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Eva Altrock
- Max-Planck Institute for Biochemistry, Martinsried, Germany.,Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Sarah Schott
- Department of Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, University of Dresden, Dresden, Germany
| | - Theresa Link
- Department of Gynecology and Obstetrics, University of Dresden, Dresden, Germany
| | - Jan Dominik Kuhlmann
- Department of Gynecology and Obstetrics, University of Dresden, Dresden, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tuebingen, Tuebingen, Germany
| | | | | | - Markus Rojewski
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Mannheim, Germany
| | - Inaam A Nakchbandi
- Max-Planck Institute for Biochemistry, Martinsried, Germany. .,Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
26
|
Sens C, Huck K, Pettera S, Uebel S, Wabnitz G, Moser M, Nakchbandi IA. Fibronectins containing extradomain A or B enhance osteoblast differentiation via distinct integrins. J Biol Chem 2017; 292:7745-7760. [PMID: 28325836 DOI: 10.1074/jbc.m116.739987] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
Fibronectin is a multidomain protein secreted by various cell types. It forms a network of fibers within the extracellular matrix and impacts intracellular processes by binding to various molecules, primarily integrin receptors on the cells. Both the presence of several isoforms and the ability of the various domains and isoforms to bind to a variety of integrins result in a wide range of effects. In vivo findings suggest that fibronectin isoforms produced by the osteoblasts enhance their differentiation. Here we report that the isoform characterized by the presence of extradomain A activates α4β1 integrin and augments osteoblast differentiation. In addition, the isoform containing extradomain B enhances the binding of fibronectin through the RGD sequence to β3-containing integrin, resulting in increased mineralization by and differentiation of osteoblasts. Our study thus reveals novel functions for two fibronectin isoforms and the mediating receptors in osteoblast differentiation.
Collapse
Affiliation(s)
- Carla Sens
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and.,the Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Katrin Huck
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and.,the Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Pettera
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and
| | - Stephan Uebel
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and
| | - Guido Wabnitz
- the Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Markus Moser
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and
| | - Inaam A Nakchbandi
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and .,the Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
27
|
Wang JP, Hielscher A. Fibronectin: How Its Aberrant Expression in Tumors May Improve Therapeutic Targeting. J Cancer 2017; 8:674-682. [PMID: 28367247 PMCID: PMC5370511 DOI: 10.7150/jca.16901] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/23/2016] [Indexed: 11/24/2022] Open
Abstract
Fibronectin is a matrix glycoprotein which has not only been found to be over-expressed in several cancers, but has been shown to participate in several steps of tumorigenesis. The purpose of this review is to illustrate how aberrant fibronectin expression influences tumor growth, invasion, metastasis and therapy resistance. In particular, this review will focus on the interactions between cell receptor ligands and fibronectin and how this interaction influences downstream signaling events that aid tumor progression. This review will further discuss the possible implications of therapeutic drugs directed against fibronectin and/or cellular interactions with fibronectin and will additionally discuss novel approaches by which to limit intra- and extra-tumoral fibronectin expression and the cellular events which lead to aberrant fibronectin expression. It is anticipated that these studies will set a basis for future research that will not only aid understanding of fibronectin and its prognostic significance, but will further elucidate novel targets for therapeutics.
Collapse
Affiliation(s)
- Jennifer Peyling Wang
- Department of Biomedical Sciences, Georgia-Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, USA
| | - Abigail Hielscher
- Department of Biomedical Sciences, Georgia-Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, USA
| |
Collapse
|