1
|
Bamberg EE, Maslanka M, Vinod-Paul K, Sams S, Pollack E, Conklin M, Kabos P, Hansen KC. Obesity-driven changes in breast tissue exhibit a pro-angiogenic extracellular matrix signature. Matrix Biol Plus 2024; 24:100162. [PMID: 39380725 PMCID: PMC11460480 DOI: 10.1016/j.mbplus.2024.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Obesity has reached epidemic proportions in the United States, emerging as a risk factor for the onset of breast cancer and a harbinger of unfavorable outcomes [1], [2], [3]. Despite limited understanding of the precise mechanisms, both obesity and breast cancer are associated with extracellular matrix (ECM) rewiring [4], [5], [6]. Utilizing total breast tissue proteomics, we analyzed normal-weight (18.5 to < 25 kg/m2), overweight (25 to < 30 kg/m2), and obese (≥30 kg/m2) individuals to identify potential ECM modifying proteins for cancer development and acceleration. Obese individuals exhibited substantial ECM alterations, marked by increased basement membrane deposition, angiogenic signatures, and ECM-modifying proteins. Notably, the collagen IV crosslinking enzyme peroxidasin (PXDN) emerged as a potential mediator of the ECM changes in individuals with an elevated body mass index (BMI), strongly correlating with angiogenic and basement membrane signatures. Furthermore, glycan-binding proteins galectin-1 (LGALS1) and galectin-3 (LGALS3), which play crucial roles in matrix interactions and angiogenesis, also strongly correlate with ECM modifications. In breast cancer, elevated PXDN, LGALS1, and LGALS3 correlate with reduced relapse-free and distant-metastatic-free survival. These proteins were significantly associated with mesenchymal stromal cell markers, indicating adipocytes and fibroblasts may be the primary contributors of the obesity-related ECM changes. Our findings unveil a pro-angiogenic ECM signature in obese breast tissue, offering potential targets to inhibit breast cancer development and progression.
Collapse
Affiliation(s)
- Ellen E. Bamberg
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mark Maslanka
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kiran Vinod-Paul
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sharon Sams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Erica Pollack
- Department of Radiology and Medical Imaging, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew Conklin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, Carbone Cancer Center (Tumor Microenvironment Program), University of Wisconsin, Madison, WI, USA
- Laboratory for Optical and Computations Instrumentation, Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Peter Kabos
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
2
|
Lengyel M, Molnár Á, Nagy T, Jdeed S, Garai I, Horváth Z, Uray IP. Zymogen granule protein 16B (ZG16B) is a druggable epigenetic target to modulate the mammary extracellular matrix. Cancer Sci 2024. [PMID: 39489500 DOI: 10.1111/cas.16382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/19/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
High tissue density of the mammary gland is considered a pro-tumorigenic factor, hence suppressing the stimuli that induce matrix buildup carries the potential for cancer interception. We found that in non-malignant mammary epithelial cells the combination of the chemopreventive agents bexarotene (Bex) and carvedilol (Carv) suppresses the zymogen granule protein 16B (ZG16B, PAUF) through an interaction of ARID1A with a proximal enhancer. Bex + Carv also reduced ZG16B levels in vivo in normal breast tissue and MDA-MB231 tumor xenografts. The relevance of ZG16B is underscored by ongoing clinical trials targeting ZG16B in pancreatic cancers, but its role in breast cancer development is unclear. In immortalized mammary epithelial cells, secreted recombinant ZG16B stimulated mitogenic kinase phosphorylation, detachment and mesenchymal characteristics, and promoted proliferation, motility and clonogenic growth. Highly concerted induction of specific laminin, collagen and integrin isoforms indicated a shift in matrix properties toward increased density and cell-matrix interactions. Exogenous ZG16B alone blocked Bex + Carv-mediated control of cell growth and migration, and antagonized Bex + Carv-induced gene programs regulating cell adhesion and migration. In breast cancer cells ZG16B induced colony formation and anchorage-independent growth, and stimulated migration in a PI3K/Akt-dependent manner. In contrast, Bex + Carv inhibited colony formation, reduced Ki67 levels, ZG16B expression and glucose uptake in MDA-MB231 xenografts. These data establish ZG16B as a druggable pro-tumorigenic target in breast cell transformation and suggest a key role of the matrisome network in rexinoid-dependent antitumor activity.
Collapse
Affiliation(s)
- Máté Lengyel
- Department of Clinical Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- The Molecular Cell and Immune Biology Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Ádám Molnár
- Department of Clinical Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Nagy
- Department of Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - Sham Jdeed
- Department of Clinical Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- The Molecular Cell and Immune Biology Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Ildikó Garai
- Department of Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Horváth
- Center of Oncoradiology, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| | - Iván P Uray
- Department of Clinical Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- The Molecular Cell and Immune Biology Doctoral School, University of Debrecen, Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
3
|
Li C, Kaur A, Pavlidaki A, Spenlé C, Rajnpreht I, Donnadieu E, Salomé N, Molitor A, Carapito R, Wack F, Erne W, Lefebvre O, Averous G, Mitrentsi I, Loustau T, Orend G. Targeting the MAtrix REgulating MOtif abolishes several hallmarks of cancer, triggering antitumor immunity. Proc Natl Acad Sci U S A 2024; 121:e2404485121. [PMID: 39382998 PMCID: PMC11494334 DOI: 10.1073/pnas.2404485121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 08/26/2024] [Indexed: 10/11/2024] Open
Abstract
Tumor-targeted therapies have often been inefficient due to the lack of concomitant control over the tumor microenvironment. Using an immunocompetent autologous breast cancer model, we investigated a MAtrix REgulating MOtif (MAREMO)-mimicking peptide, which inhibits the protumorigenic extracellular matrix (ECM) molecule tenascin-C that activates several cancer hallmarks. In cultured cells, targeting the MAREMO blocks tenascin-C signaling involved in cell adhesion and immune-suppression by inhibiting tenascin-C interactions with fibronectin, TGFβ, CXCL12, and others, thereby blocking downstream events. Using RNASequencing and various genetic, molecular, in situ, and in vivo assays, we demonstrate that the MAREMO peptide similarly blocks multiple tenascin-C functions in vivo. This includes releasing tumor-infiltrating leukocytes, including CD8+ T cells, from the stroma. The MAREMO peptide also triggers interferon signaling, restoring antitumor immunity, contributing to tumor growth inhibition and reduced dissemination. The MAREMO peptide targets tumor cells directly by promoting growth suppression and inhibiting phenotypic plasticity, subsequently enhancing responsiveness to the endogenous death inducer tumor necrosis factor-related apoptosis-inducing ligand, as shown by a loss-of-function approach. Moreover, the MAREMO peptide largely subdues the tumor bed by depleting fibroblasts, repressing tenascin-C and other ECM molecules, and restoring the function of the few remaining blood vessels. In conclusion, targeting tenascin-C with a MAREMO peptide represents a powerful anticancer strategy with a broad inhibition of several cancer hallmarks.
Collapse
Affiliation(s)
- Chengbei Li
- University of Strasbourg, Strasbourg67091, France
- INSERM U1109, The Tumor Microenvironment Laboratory, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
| | - Amanpreet Kaur
- University of Strasbourg, Strasbourg67091, France
- INSERM U1109, The Tumor Microenvironment Laboratory, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
| | - Alexia Pavlidaki
- University of Strasbourg, Strasbourg67091, France
- INSERM U1109, The Tumor Microenvironment Laboratory, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
| | - Caroline Spenlé
- École Supérieure de Biotechnologie de Strasbourg (ESBS) UMR 7242, Groupe Peptide Thérapeutique, University of Strasbourg, Illkirch67400, France
| | - Irena Rajnpreht
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Equipe Labellisée Ligue Contre le Cancer, Paris75014, France
| | - Emmanuel Donnadieu
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Equipe Labellisée Ligue Contre le Cancer, Paris75014, France
| | - Nathalie Salomé
- University of Strasbourg, Strasbourg67091, France
- INSERM U1109, The Tumor Microenvironment Laboratory, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
| | - Anne Molitor
- University of Strasbourg, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, Plateforme GENOMAX, Institut Thématique Interdisciplinaire de Médecine de Précision de Strasbourg, Transplantex Next Generation (NG), Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg67091, France
| | - Raphael Carapito
- University of Strasbourg, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, Plateforme GENOMAX, Institut Thématique Interdisciplinaire de Médecine de Précision de Strasbourg, Transplantex Next Generation (NG), Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg67091, France
| | - Fanny Wack
- University of Strasbourg, Strasbourg67091, France
- INSERM U1109, The Tumor Microenvironment Laboratory, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
| | - William Erne
- University of Strasbourg, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
- INSERM U1109, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy Laboratory, Hautepierre, Strasbourg67091, France
| | - Olivier Lefebvre
- University of Strasbourg, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
- INSERM U1109, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy Laboratory, Hautepierre, Strasbourg67091, France
| | - Gerlinde Averous
- Département de Pathologie, University Hospital Strasbourg, Strasbourg67200, France
| | - Ioanna Mitrentsi
- University of Strasbourg, Strasbourg67091, France
- INSERM U1109, The Tumor Microenvironment Laboratory, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
| | - Thomas Loustau
- University of Strasbourg, Strasbourg67091, France
- INSERM U1109, The Tumor Microenvironment Laboratory, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
- University of Strasbourg, Institut Universitaire Technologique (IUT) Louis Pasteur, Schiltigheim67300, France
| | - Gertraud Orend
- University of Strasbourg, Strasbourg67091, France
- INSERM U1109, The Tumor Microenvironment Laboratory, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Strasbourg67091, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg67091, France
- INSERM U1109, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy Laboratory, Hautepierre, Strasbourg67091, France
| |
Collapse
|
4
|
Arpinati L, Carradori G, Scherz-Shouval R. CAF-induced physical constraints controlling T cell state and localization in solid tumours. Nat Rev Cancer 2024; 24:676-693. [PMID: 39251836 DOI: 10.1038/s41568-024-00740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Solid tumours comprise cancer cells that engage in continuous interactions with non-malignant cells and with acellular components, forming the tumour microenvironment (TME). The TME has crucial and diverse roles in tumour progression and metastasis, and substantial efforts have been dedicated into understanding the functions of different cell types within the TME. These efforts highlighted the importance of non-cell-autonomous signalling in cancer, mediating interactions between the cancer cells, the immune microenvironment and the non-immune stroma. Much of this non-cell-autonomous signalling is mediated through acellular components of the TME, known as the extracellular matrix (ECM), and controlled by the cells that secrete and remodel the ECM - the cancer-associated fibroblasts (CAFs). In this Review, we delve into the complex crosstalk among cancer cells, CAFs and immune cells, highlighting the effects of CAF-induced ECM remodelling on T cell functions and offering insights into the potential of targeting ECM components to improve cancer therapies.
Collapse
Affiliation(s)
- Ludovica Arpinati
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Giulia Carradori
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
5
|
Barros da Silva P, Zhao X, Bidarra SJ, Nascimento DS, LaLone V, Lourenço BN, Paredes J, Stevens MM, Barrias CC. Tunable Hybrid Hydrogels of Alginate and Cell-Derived dECM to Study the Impact of Matrix Alterations on Epithelial-to-Mesenchymal Transition. Adv Healthc Mater 2024:e2401032. [PMID: 39246099 DOI: 10.1002/adhm.202401032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/27/2024] [Indexed: 09/10/2024]
Abstract
Epithelial-to-mesenchymal transition (EMT) is crucial for tumor progression, being linked to alterations in the extracellular matrix (ECM). Understanding the ECM's role in EMT can uncover new therapeutic targets, yet replicating these interactions in vitro remains challenging. It is shown that hybrid hydrogels of alginate (ALG) and cell-derived decellularized ECM (dECM), with independently tunable composition and stiffness, are useful 3D-models to explore the impact of the breast tumor matrix on EMT. Soft RGD-ALG hydrogels (200 Pa), used as neutral bulk material, supported mammary epithelial cells morphogenesis without spontaneous EMT, allowing to define the gene, protein, and biochemical profiles of cells at different TGFβ1-induced EMT states. To mimic the breast tumor composition, dECM from TGFβ1-activated fibroblasts (adECM) are generated, which shows upregulation of tumor-associated proteins compared to ndECM from normal fibroblasts. Using hybrid adECM-ALG hydrogels, it is shown that the presence of adECM induces partial EMT in normal epithelial cells, and amplifes TGF-β1 effects compared to ALG and ndECM-ALG. Increasing the hydrogel stiffness to tumor-like levels (2.5 kPa) have a synergistic effect, promoting a more evident EMT. These findings shed light on the complex interplay between matrix composition and stiffness in EMT, underscoring the utility of dECM-ALG hydrogels as a valuable in vitro platform for cancer research.
Collapse
Affiliation(s)
- P Barros da Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, 4200-135, Portugal
| | - Xiaoyu Zhao
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Sílvia J Bidarra
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
| | - Diana S Nascimento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4200-135, Portugal
| | - Vernon LaLone
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Department of Materials, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Bianca N Lourenço
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, 4200-135, Portugal
| | - Joana Paredes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- FMUP - Faculdade de Medicina da Universidade do Porto, Porto, 4200-319, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, 4200-135, Portugal
| | - Molly M Stevens
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Department of Materials, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - C C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4200-135, Portugal
| |
Collapse
|
6
|
Hong J, Jin HJ, Choi MR, Lim DWT, Park JE, Kim YS, Lim SB. Matrisomics: Beyond the extracellular matrix for unveiling tumor microenvironment. Biochim Biophys Acta Rev Cancer 2024; 1879:189178. [PMID: 39241895 DOI: 10.1016/j.bbcan.2024.189178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
The matrisome, a group of proteins constituting or interacting with the extracellular matrix (ECM), has garnered attention as a potent regulator of cancer progression. An increasing number of studies have focused on cancer matrisome utilizing diverse -omics approaches. Here, we present diverse patterns of matrisomal populations within cancer tissues, exploring recent -omics studies spanning different '-omics' levels (epigenomics, genomics, transcriptomics, and proteomics), as well as newly developed sequencing techniques such as single-cell RNA sequencing and spatial transcriptomics. Some matrisome genes showed uniform patterns of upregulated or downregulated expression across various cancers, while others displayed different expression patterns according to the cancer types. This matrisomal dysregulation in cancer was further examined according to their originating cell type and spatial location in the tumor tissue. Experimental studies were also collected to demonstrate the identified roles of matrisome genes during cancer progression. Interestingly, many studies on cancer matrisome have suggested matrisome genes as effective biomarkers in cancer research. Although the specific mechanisms and clinical applications of cancer matrisome have not yet been fully elucidated, recent techniques and analyses on cancer matrisomics have emphasized their biological importance in cancer progression and their clinical implications in deciding the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Jiwon Hong
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Hyo Joon Jin
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Mi Ran Choi
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Darren Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre, Singapore 168583, Singapore
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - You-Sun Kim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
7
|
Zhou L, Li G, Yao J, Wang J, Yao X, Ye Z, Zheng D, Song K, Zhang H, Zhang X, Shuai J, Ye F, Li M, Li Y, Chen G, Cheng Y, Liu H, Shaw P, Liu L. Emulation and evaluation of tumor cell combined chemotherapy in isotropic/anisotropic collagen fiber microenvironments. LAB ON A CHIP 2024; 24:2999-3014. [PMID: 38742451 DOI: 10.1039/d4lc00051j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The rapid emergence of anisotropic collagen fibers in the tissue microenvironment is a critical transition point in late-stage breast cancer. Specifically, the fiber orientation facilitates the likelihood of high-speed tumor cell invasion and metastasis, which pose lethal threats to patients. Thus, based on this transition point, one key issue is how to determine and evaluate efficient combination chemotherapy treatments in late-stage cancer. In this study, we designed a collagen microarray chip containing 241 high-throughput microchambers with embedded metastatic breast cancer cell MDA-MB-231-RFP. By utilizing collagen's unique structure and hydromechanical properties, the chip constructed three-dimensional isotropic and anisotropic collagen fiber structures to emulate the tumor cell microenvironment at early and late stages. We injected different chemotherapeutic drugs into its four channels and obtained composite biochemical concentration profiles. Our results demonstrate that anisotropic collagen fibers promote cell proliferation and migration more than isotropic collagen fibers, suggesting that the geometric arrangement of fibers plays an important role in regulating cell behavior. Moreover, the presence of anisotropic collagen fibers may be a potential factor leading to the poor efficacy of combined chemotherapy in late-stage breast cancer. We investigated the efficacy of various chemotherapy drugs using cell proliferation inhibitors paclitaxel and gemcitabine and tumor cell migration inhibitors 7rh and PP2. To ensure the validity of our findings, we followed a systematic approach that involved testing the inhibitory effects of these drugs. According to our results, the drug combinations' effectiveness could be ordered as follows: paclitaxel + gemcitabine > gemcitabine + 7rh > PP2 + paclitaxel > 7rh + PP2. This study shows that the biomimetic chip system not only facilitates the creation of a realistic in vitro model for examining the cell migration mechanism in late-stage breast cancer but also has the potential to function as an effective tool for future chemotherapy assessment and personalized medicine.
Collapse
Affiliation(s)
- Lianjie Zhou
- Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 401331, China.
| | - Guoqiang Li
- Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 401331, China.
- Chongqing Key Laboratory for Resource Utilization of Heavy Metal Wastewater, Chongqing University of Arts and Sciences, Yongchuan 402160, PR China
| | - Jingru Yao
- Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 401331, China.
| | - Jing Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Xiyao Yao
- Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 401331, China.
| | - Zhikai Ye
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Dongtian Zheng
- Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 401331, China.
| | - Kena Song
- College of Medical Technology and Engineering, Henan University of Science and Technology, Henan 471023, China
| | - Hongfei Zhang
- Hygeia International Cancer Hospital, Chongqing 401331, China
| | - Xianquan Zhang
- Hygeia International Cancer Hospital, Chongqing 401331, China
| | - Jianwei Shuai
- Department of Physics, Xiamen University, Xiamen 361005, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Fangfu Ye
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Ming Li
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Yufeng Li
- Shaanxi Provincial Key Laboratory of Photonics & Information Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Guo Chen
- Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 401331, China.
| | - Yuyan Cheng
- Chongqing Key Laboratory for Resource Utilization of Heavy Metal Wastewater, Chongqing University of Arts and Sciences, Yongchuan 402160, PR China
| | - He Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang 325000, China.
| | - Peter Shaw
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang 325000, China.
| | - Liyu Liu
- Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
8
|
Darula Z, McCabe MC, Barrett A, Schmitt LR, Maslanka MD, Saviola AJ, Orgel J, Burlingame A, Staab-Weijnitz CA, Stenmark K, Weaver V, Chalkley RJ, Hansen KC. Assessing Heterogeneity in the N-Telopeptides of Type I Collagen by Mass Spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.31.587441. [PMID: 38585857 PMCID: PMC10996605 DOI: 10.1101/2024.03.31.587441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Collagen cross-links created by the lysyl oxidase and lysyl hydroxylase families of enzymes are a significant contributing factor to the biomechanical strength and rigidity of tissues, which in turn influence cell signaling and ultimately cell phenotype. In the clinic, the proteolytically liberated N-terminal cross-linked peptide of collagen I (NTX) is used as a biomarker of bone and connective tissue turnover, which is altered in several disease processes. Despite the clinical utility of these collagen breakdown products, the majority of the cross-linked peptide species have not been identified in proteomic datasets. Here we evaluate several parameters for the preparation and identification of these peptides from the collagen I-rich Achilles tendon. Our refined approach involving chemical digestion for protein solubilization coupled with mass spectrometry allows for the identification of the NTX cross-links in a range of modification states. Based on the specificity of the enzymatic cross-linking reaction we utilized follow-up variable modification searches to facilitate identification with a wider range of analytical workflows. We then applied a spectral library approach to identify differences in collagen cross-links in bovine pulmonary hypertension. The presented method offers unique opportunities to understand extracellular matrix remodeling events in development, aging, wound healing, and fibrotic disease that modulate collagen architecture through lysyl-hydroxylase and lysyl-oxidase enzymes.
Collapse
|
9
|
Park H, Li B, Liu Y, Nelson MS, Wilson HM, Sifakis E, Eliceiri KW. Collagen fiber centerline tracking in fibrotic tissue via deep neural networks with variational autoencoder-based synthetic training data generation. Med Image Anal 2023; 90:102961. [PMID: 37802011 PMCID: PMC10591913 DOI: 10.1016/j.media.2023.102961] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 10/08/2023]
Abstract
The role of fibrillar collagen in the tissue microenvironment is critical in disease contexts ranging from cancers to chronic inflammations, as evidenced by many studies. Quantifying fibrillar collagen organization has become a powerful approach for characterizing the topology of collagen fibers and studying the role of collagen fibers in disease progression. We present a deep learning-based pipeline to quantify collagen fibers' topological properties in microscopy-based collagen images from pathological tissue samples. Our method leverages deep neural networks to extract collagen fiber centerlines and deep generative models to create synthetic training data, addressing the current shortage of large-scale annotations. As a part of this effort, we have created and annotated a collagen fiber centerline dataset, with the hope of facilitating further research in this field. Quantitative measurements such as fiber orientation, alignment, density, and length can be derived based on the centerline extraction results. Our pipeline comprises three stages. Initially, a variational autoencoder is trained to generate synthetic centerlines possessing controllable topological properties. Subsequently, a conditional generative adversarial network synthesizes realistic collagen fiber images from the synthetic centerlines, yielding a synthetic training set of image-centerline pairs. Finally, we train a collagen fiber centerline extraction network using both the original and synthetic data. Evaluation using collagen fiber images from pancreas, liver, and breast cancer samples collected via second-harmonic generation microscopy demonstrates our pipeline's superiority over several popular fiber centerline extraction tools. Incorporating synthetic data into training further enhances the network's generalizability. Our code is available at https://github.com/uw-loci/collagen-fiber-metrics.
Collapse
Affiliation(s)
- Hyojoon Park
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53706, USA.
| | - Bin Li
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53706, USA.
| | - Yuming Liu
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Michael S Nelson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Helen M Wilson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Eftychios Sifakis
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53706, USA.
| |
Collapse
|
10
|
Neagu AN, Whitham D, Bruno P, Morrissiey H, Darie CA, Darie CC. Omics-Based Investigations of Breast Cancer. Molecules 2023; 28:4768. [PMID: 37375323 DOI: 10.3390/molecules28124768] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer (BC) is characterized by an extensive genotypic and phenotypic heterogeneity. In-depth investigations into the molecular bases of BC phenotypes, carcinogenesis, progression, and metastasis are necessary for accurate diagnoses, prognoses, and therapy assessments in predictive, precision, and personalized oncology. This review discusses both classic as well as several novel omics fields that are involved or should be used in modern BC investigations, which may be integrated as a holistic term, onco-breastomics. Rapid and recent advances in molecular profiling strategies and analytical techniques based on high-throughput sequencing and mass spectrometry (MS) development have generated large-scale multi-omics datasets, mainly emerging from the three "big omics", based on the central dogma of molecular biology: genomics, transcriptomics, and proteomics. Metabolomics-based approaches also reflect the dynamic response of BC cells to genetic modifications. Interactomics promotes a holistic view in BC research by constructing and characterizing protein-protein interaction (PPI) networks that provide a novel hypothesis for the pathophysiological processes involved in BC progression and subtyping. The emergence of new omics- and epiomics-based multidimensional approaches provide opportunities to gain insights into BC heterogeneity and its underlying mechanisms. The three main epiomics fields (epigenomics, epitranscriptomics, and epiproteomics) are focused on the epigenetic DNA changes, RNAs modifications, and posttranslational modifications (PTMs) affecting protein functions for an in-depth understanding of cancer cell proliferation, migration, and invasion. Novel omics fields, such as epichaperomics or epimetabolomics, could investigate the modifications in the interactome induced by stressors and provide PPI changes, as well as in metabolites, as drivers of BC-causing phenotypes. Over the last years, several proteomics-derived omics, such as matrisomics, exosomics, secretomics, kinomics, phosphoproteomics, or immunomics, provided valuable data for a deep understanding of dysregulated pathways in BC cells and their tumor microenvironment (TME) or tumor immune microenvironment (TIMW). Most of these omics datasets are still assessed individually using distinct approches and do not generate the desired and expected global-integrative knowledge with applications in clinical diagnostics. However, several hyphenated omics approaches, such as proteo-genomics, proteo-transcriptomics, and phosphoproteomics-exosomics are useful for the identification of putative BC biomarkers and therapeutic targets. To develop non-invasive diagnostic tests and to discover new biomarkers for BC, classic and novel omics-based strategies allow for significant advances in blood/plasma-based omics. Salivaomics, urinomics, and milkomics appear as integrative omics that may develop a high potential for early and non-invasive diagnoses in BC. Thus, the analysis of the tumor circulome is considered a novel frontier in liquid biopsy. Omics-based investigations have applications in BC modeling, as well as accurate BC classification and subtype characterization. The future in omics-based investigations of BC may be also focused on multi-omics single-cell analyses.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Carol I Bvd, No. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Pathea Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Hailey Morrissiey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Celeste A Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Costel C Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| |
Collapse
|
11
|
Bruni S, Mercogliano MF, Mauro FL, Cordo Russo RI, Schillaci R. Cancer immune exclusion: breaking the barricade for a successful immunotherapy. Front Oncol 2023; 13:1135456. [PMID: 37284199 PMCID: PMC10239871 DOI: 10.3389/fonc.2023.1135456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Immunotherapy has changed the course of cancer treatment. The initial steps were made through tumor-specific antibodies that guided the setup of an antitumor immune response. A new and successful generation of antibodies are designed to target immune checkpoint molecules aimed to reinvigorate the antitumor immune response. The cellular counterpart is the adoptive cell therapy, where specific immune cells are expanded or engineered to target cancer cells. In all cases, the key for achieving positive clinical resolutions rests upon the access of immune cells to the tumor. In this review, we focus on how the tumor microenvironment architecture, including stromal cells, immunosuppressive cells and extracellular matrix, protects tumor cells from an immune attack leading to immunotherapy resistance, and on the available strategies to tackle immune evasion.
Collapse
|
12
|
Neagu AN, Whitham D, Seymour L, Haaker N, Pelkey I, Darie CC. Proteomics-Based Identification of Dysregulated Proteins and Biomarker Discovery in Invasive Ductal Carcinoma, the Most Common Breast Cancer Subtype. Proteomes 2023; 11:13. [PMID: 37092454 PMCID: PMC10123686 DOI: 10.3390/proteomes11020013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Invasive ductal carcinoma (IDC) is the most common histological subtype of malignant breast cancer (BC), and accounts for 70-80% of all invasive BCs. IDC demonstrates great heterogeneity in clinical and histopathological characteristics, prognoses, treatment strategies, gene expressions, and proteomic profiles. Significant proteomic determinants of the progression from intraductal pre-invasive malignant lesions of the breast, which characterize a ductal carcinoma in situ (DCIS), to IDC, are still poorly identified, validated, and clinically applied. In the era of "6P" medicine, it remains a great challenge to determine which patients should be over-treated versus which need to be actively monitored without aggressive treatment. The major difficulties for designating DCIS to IDC progression may be solved by understanding the integrated genomic, transcriptomic, and proteomic bases of invasion. In this review, we showed that multiple proteomics-based techniques, such as LC-MS/MS, MALDI-ToF MS, SELDI-ToF-MS, MALDI-ToF/ToF MS, MALDI-MSI or MasSpec Pen, applied to in-tissue, off-tissue, BC cell lines and liquid biopsies, improve the diagnosis of IDC, as well as its prognosis and treatment monitoring. Classic proteomics strategies that allow the identification of dysregulated protein expressions, biological processes, and interrelated pathway analyses based on aberrant protein-protein interaction (PPI) networks have been improved to perform non-invasive/minimally invasive biomarker detection of early-stage IDC. Thus, in modern surgical oncology, highly sensitive, rapid, and accurate MS-based detection has been coupled with "proteome point sampling" methods that allow for proteomic profiling by in vivo "proteome point characterization", or by minimal tissue removal, for ex vivo accurate differentiation and delimitation of IDC. For the detection of low-molecular-weight proteins and protein fragments in bodily fluids, LC-MS/MS and MALDI-MS techniques may be coupled to enrich and capture methods which allow for the identification of early-stage IDC protein biomarkers that were previously invisible for MS-based techniques. Moreover, the detection and characterization of protein isoforms, including posttranslational modifications of proteins (PTMs), is also essential to emphasize specific molecular mechanisms, and to assure the early-stage detection of IDC of the breast.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Carol I bvd. No. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Logan Seymour
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Norman Haaker
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Isabella Pelkey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| |
Collapse
|
13
|
Benn MC, Pot SA, Moeller J, Yamashita T, Fonta CM, Orend G, Kollmannsberger P, Vogel V. How the mechanobiology orchestrates the iterative and reciprocal ECM-cell cross-talk that drives microtissue growth. SCIENCE ADVANCES 2023; 9:eadd9275. [PMID: 36989370 PMCID: PMC10058249 DOI: 10.1126/sciadv.add9275] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Controlled tissue growth is essential for multicellular life and requires tight spatiotemporal control over cell proliferation and differentiation until reaching homeostasis. As cells synthesize and remodel extracellular matrix, tissue growth processes can only be understood if the reciprocal feedback between cells and their environment is revealed. Using de novo-grown microtissues, we identified crucial actors of the mechanoregulated events, which iteratively orchestrate a sharp transition from tissue growth to maturation, requiring a myofibroblast-to-fibroblast transition. Cellular decision-making occurs when fibronectin fiber tension switches from highly stretched to relaxed, and it requires the transiently up-regulated appearance of tenascin-C and tissue transglutaminase, matrix metalloprotease activity, as well as a switch from α5β1 to α2β1 integrin engagement and epidermal growth factor receptor signaling. As myofibroblasts are associated with wound healing and inflammatory or fibrotic diseases, crucial knowledge needed to advance regenerative strategies or to counter fibrosis and cancer progression has been gained.
Collapse
Affiliation(s)
- Mario C. Benn
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Simon A. Pot
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Jens Moeller
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Tadahiro Yamashita
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Charlotte M. Fonta
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France
- Université Strasbourg, Strasbourg 67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Philip Kollmannsberger
- Biomedical Physics, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, Düsseldorf 40225, Germany
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| |
Collapse
|
14
|
Naba A. 10 years of extracellular matrix proteomics: Accomplishments, challenges, and future perspectives. Mol Cell Proteomics 2023; 22:100528. [PMID: 36918099 PMCID: PMC10152135 DOI: 10.1016/j.mcpro.2023.100528] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023] Open
Abstract
The extracellular matrix (ECM) is a complex assembly of hundreds of proteins forming the architectural scaffold of multicellular organisms. In addition to its structural role, the ECM conveys signals orchestrating cellular phenotypes. Alterations of ECM composition, abundance, structure, or mechanics, have been linked to diseases and disorders affecting all physiological systems, including fibrosis and cancer. Deciphering the protein composition of the ECM and how it changes in pathophysiological contexts is thus the first step toward understanding the roles of the ECM in health and disease and toward the development of therapeutic strategies to correct disease-causing ECM alterations. Potentially, the ECM also represents a vast, yet untapped reservoir of disease biomarkers. ECM proteins are characterized by unique biochemical properties that have hindered their study: they are large, heavily and uniquely post-translationally modified, and highly insoluble. Overcoming these challenges, we and others have devised mass-spectrometry-based proteomic approaches to define the ECM composition, or "matrisome", of tissues. This review provides a historical overview of ECM proteomics research and presents the latest advances that now allow the profiling of the ECM of healthy and diseased tissues. The second part highlights recent examples illustrating how ECM proteomics has emerged as a powerful discovery pipeline to identify prognostic cancer biomarkers. The third part discusses remaining challenges limiting our ability to translate findings to clinical application and proposes approaches to overcome them. Last, the review introduces readers to resources available to facilitate the interpretation of ECM proteomics datasets. The ECM was once thought to be impenetrable. MS-based proteomics has proven to be a powerful tool to decode the ECM. In light of the progress made over the past decade, there are reasons to believe that the in-depth exploration of the matrisome is within reach and that we may soon witness the first translational application of ECM proteomics.
Collapse
Affiliation(s)
- Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Cancer Center, Chicago, IL 60612, USA.
| |
Collapse
|
15
|
Fonta CM, Loustau T, Li C, Poilil Surendran S, Hansen U, Murdamoothoo D, Benn MC, Velazquez-Quesada I, Carapito R, Orend G, Vogel V. Infiltrating CD8+ T cells and M2 macrophages are retained in tumor matrix tracks enriched in low tension fibronectin fibers. Matrix Biol 2023; 116:1-27. [PMID: 36669744 DOI: 10.1016/j.matbio.2023.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 12/31/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
Tracks rich in matrix and cells, as described in several cancer types, have immunosuppressive functions and separate tumor nests and stroma, yet their origin is unknown. Immunostainings of cryosections from mouse breast tumors show that these tracks are bordered by an endothelial-like basement membrane, filled with fibers of collagen adjacent to tenascin-C (TNC) and low-tension fibronectin (Fn) fibers. While present in early-stage tumors and maturing with time, tracks still form under TNC KO conditions, however, host (not tumor cell)-derived TNC is important for track maturation. Tumor infiltrating leukocytes (mostly M2 macrophages and CD8+ T cells) are retained in tracks of early-stage tumors. Following track maturation, retained tumor infiltrating leukocyte (TIL) numbers get reduced and more CD8+ TIL enter the tumor nests in the absence of TNC. As these tracks are enriched with platelets and fibrinogen and have a demarcating endothelial-like basement membrane often adjacent to endothelial cells, this suggests a role of blood vessels in the formation of these tracks. The Fn fiber tension probe FnBPA5 colocalizes with TNC and immune cells in the tracks and shows decreased binding in tracks lacking TNC. Consequently, FnBPA5 can serve as probe for tumor matrix tracks that have immune suppressive properties.
Collapse
Affiliation(s)
- Charlotte M Fonta
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Vladimir Prelog Weg, Zurich CH-8093, Switzerland
| | - Thomas Loustau
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d' Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France; Université Strasbourg, Strasbourg 67000, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Chengbei Li
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d' Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France; Université Strasbourg, Strasbourg 67000, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Suchithra Poilil Surendran
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d' Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France; Université Strasbourg, Strasbourg 67000, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine (IMM), University Hospital Muenster, Muenster, Federal Republic of Germany
| | - Devadarssen Murdamoothoo
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d' Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France; MN3T (The Microenvironmental Niche in Tumorigenesis and Targeted Therapy), INSERM U1109, 3 avenue Molière, Strasbourg, Hautepierre, France; Université Strasbourg, Strasbourg 67000, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Mario C Benn
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Vladimir Prelog Weg, Zurich CH-8093, Switzerland
| | - Ines Velazquez-Quesada
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d' Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France; MN3T (The Microenvironmental Niche in Tumorigenesis and Targeted Therapy), INSERM U1109, 3 avenue Molière, Strasbourg, Hautepierre, France; Université Strasbourg, Strasbourg 67000, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Raphael Carapito
- Université Strasbourg, Strasbourg 67000, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France; Platform GENOMAX, INSERM UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, LabEx TRANSPLANTEX, Strasbourg 67091, France
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d' Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France; MN3T (The Microenvironmental Niche in Tumorigenesis and Targeted Therapy), INSERM U1109, 3 avenue Molière, Strasbourg, Hautepierre, France; Université Strasbourg, Strasbourg 67000, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France.
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Vladimir Prelog Weg, Zurich CH-8093, Switzerland.
| |
Collapse
|
16
|
K Patel K, Hassan D, Nair S, Tejovath S, Kahlon SS, Peddemul A, Sikandar R, Mostafa JA. Role of Immunotherapy in the Treatment of Triple-Negative Breast Cancer: A Literature Review. Cureus 2022; 14:e31729. [PMID: 36569674 PMCID: PMC9771573 DOI: 10.7759/cureus.31729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022] Open
Abstract
Numerous malignancies, including metastatic triple-negative breast cancer (TNBC), which has long been associated with a poor prognosis, have been transformed by the widespread use of immunotherapy. Immune checkpoint inhibitors (ICIs) that target and block programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1) have demonstrated encouraging outcomes in the treatment of patients with metastatic TNBC. The PD-1 inhibitor pembrolizumab is the first-line treatment of metastatic PD-L1+ TNBC in combination with chemotherapy, and the PD-L1 inhibitor atezolizumab has also shown clinical activity. The median progression-free survival for pembrolizumab or atezolizumab combined with chemotherapy increased by 4.1 months and 2.5 months, respectively, with the addition of immunotherapy. Despite this progress, there is still more to be desired. The addition of immunotherapy to chemotherapy improved the pathological complete response (PCR) rate compared to chemotherapy with placebo in landmark phase III trials in the early-stage neoadjuvant context, whereas others reported no meaningful improvement in PCR. There are various ongoing trials that show that more research and studies are needed for components in the TNBC microenvironment and to further explore its importance in the treatment of TNBC.
Collapse
Affiliation(s)
- Khushbu K Patel
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Danial Hassan
- Health Care Profession, Ministry of Public Health, Doha, QAT
- Cardiology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shaalina Nair
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sreedevi Tejovath
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Simranjit S Kahlon
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aishwarya Peddemul
- Obstetrics and Gynecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Rabia Sikandar
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jihan A Mostafa
- Psychiatry, Professional Psychotherapy, Cognitive Behavioral Psychotherapy, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
17
|
Yamada KM, Doyle AD, Lu J. Cell-3D matrix interactions: recent advances and opportunities. Trends Cell Biol 2022; 32:883-895. [PMID: 35410820 PMCID: PMC9464680 DOI: 10.1016/j.tcb.2022.03.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/03/2023]
Abstract
Tissues consist of cells and their surrounding extracellular matrix (ECM). Cell-ECM interactions play crucial roles in embryonic development, differentiation, tissue remodeling, and diseases including fibrosis and cancer. Recent research advances in characterizing cell-matrix interactions include detailed descriptions of hundreds of ECM and associated molecules, their complex intermolecular interactions in development and disease, identification of distinctive modes of cell migration in different 3D ECMs, and new insights into mechanisms of organ formation. Exploring the roles of the physical features of different ECM microenvironments and the bidirectional regulation of cell signaling and matrix organization emphasize the dynamic nature of these interactions, which can include feedback loops that exacerbate disease. Understanding mechanisms of cell-matrix interactions can potentially lead to targeted therapeutic interventions.
Collapse
Affiliation(s)
- Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Andrew D Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jiaoyang Lu
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Lee HJ, Gari MK, Inman DR, Rosenkrans ZT, Burkel BM, Olson AP, Engle JW, Hernandez R, Ponik SM, Kwon GS. Multimodal imaging demonstrates enhanced tumor exposure of PEGylated FUD peptide in breast cancer. J Control Release 2022; 350:284-297. [PMID: 35995299 PMCID: PMC9841600 DOI: 10.1016/j.jconrel.2022.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/01/2022] [Accepted: 08/13/2022] [Indexed: 01/23/2023]
Abstract
In breast cancer, the extracellular matrix (ECM) undergoes remodeling and changes the tumor microenvironment to support tumor progression and metastasis. Fibronectin (FN) assembly is an important step in the regulation of the tumor microenvironment since the FN matrix precedes the deposition of various other ECM proteins, controls immune cell infiltration, and serves as a reservoir for cytokines and growth factors. Therefore, FN is an attractive target for breast cancer therapy and imaging. Functional Upstream Domain (FUD) is a 6-kDa peptide targeting the N-terminal 70-kDa domain of FN, which is critical for fibrillogenesis. FUD has previously been shown to function as an anti-fibrotic peptide both in vitro and in vivo. In this work, we conjugated the FUD peptide with 20-kDa of PEG (PEG-FUD) and demonstrated its improved tumor exposure compared to non-PEGylated FUD in a murine breast cancer model via multiple imaging modalities. Importantly, PEG-FUD peptide retained a nanomolar binding affinity for FN and maintained in vitro plasma stability for up to 48 h. Cy5-labeled PEG-FUD bound to exogenous or endogenous FN assembled by fibroblasts. The in vivo fluorescence imaging with Cy5-labeled FUD and FUD conjugates demonstrated that PEGylation of the FUD peptide enhanced blood exposure after subcutaneous (SC) injection and significantly increased accumulation of FUD peptide in 4T1 mammary tumors. Intravital microscopy confirmed that Cy5-labeled PEG-FUD deposited mostly in the extravascular region of the tumor microenvironment after SC administration. Lastly, positron emission tomography/computed tomography imaging showed that 64Cu-labeled PEG-FUD preferentially accumulated in the 4T1 tumors with improved tumor uptake compared to 64Cu-labeled FUD (48 h: 1.35 ± 0.05 vs. 0.59 ± 0.03 %IA/g, P < 0.001) when injected intravenously (IV). The results indicate that PEG-FUD targets 4T1 breast cancer with enhanced tumor retention compared to non-PEGylated FUD, and biodistribution profiles of PEG-FUD after SC and IV injection may guide the optimization of PEG-FUD as a therapeutic and/or imaging agent for use in vivo.
Collapse
Affiliation(s)
- Hye Jin Lee
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, Wisconsin, 53705, USA
| | - Metti K. Gari
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, 1111 Highland Avenue, WIMRII, Madison, Wisconsin, 53705, USA
| | - David R. Inman
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, 1111 Highland Avenue, WIMRII, Madison, Wisconsin, 53705, USA
| | - Zachary T. Rosenkrans
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, Wisconsin, 53705, USA
| | - Brian M. Burkel
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, 1111 Highland Avenue, WIMRII, Madison, Wisconsin, 53705, USA
| | - Aeli P. Olson
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, Wisconsin, 53705, USA
| | - Jonathan W. Engle
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, Wisconsin, 53705, USA
| | - Reinier Hernandez
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, Wisconsin, 53705, USA,Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, Wisconsin, 53705, USA,Carbone Cancer Center, University of Wisconsin - Madison, Madison, Wisconsin, 53705, USA
| | - Suzanne M. Ponik
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, 1111 Highland Avenue, WIMRII, Madison, Wisconsin, 53705, USA,Carbone Cancer Center, University of Wisconsin - Madison, Madison, Wisconsin, 53705, USA
| | - Glen S. Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, Wisconsin, 53705, USA,Carbone Cancer Center, University of Wisconsin - Madison, Madison, Wisconsin, 53705, USA
| |
Collapse
|
19
|
Yilmaz A, Loustau T, Salomé N, Poilil Surendran S, Li C, Tucker RP, Izzi V, Lamba R, Koch M, Orend G. Advances on the roles of tenascin-C in cancer. J Cell Sci 2022; 135:276631. [PMID: 36102918 PMCID: PMC9584351 DOI: 10.1242/jcs.260244] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The roles of the extracellular matrix molecule tenascin-C (TNC) in health and disease have been extensively reviewed since its discovery over 40 years ago. Here, we will describe recent insights into the roles of TNC in tumorigenesis, angiogenesis, immunity and metastasis. In addition to high levels of expression in tumors, and during chronic inflammation, and bacterial and viral infection, TNC is also expressed in lymphoid organs. This supports potential roles for TNC in immunity control. Advances using murine models with engineered TNC levels were instrumental in the discovery of important functions of TNC as a danger-associated molecular pattern (DAMP) molecule in tissue repair and revealed multiple TNC actions in tumor progression. TNC acts through distinct mechanisms on many different cell types with immune cells coming into focus as important targets of TNC in cancer. We will describe how this knowledge could be exploited for cancer disease management, in particular for immune (checkpoint) therapies.
Collapse
Affiliation(s)
- Alev Yilmaz
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Thomas Loustau
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Nathalie Salomé
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Suchithra Poilil Surendran
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Chengbei Li
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Richard P. Tucker
- University of California at Davis 4 Department of Cell Biology and Human Anatomy , , 95616 Davis, CA , USA
| | - Valerio Izzi
- University of Oulu 5 Faculty of Biochemistry and Molecular Medicine , , FI-90014 Oulu , Finland
- University of Oulu 6 Faculty of Medicine , , FI-90014 Oulu , Finland
| | - Rijuta Lamba
- University of Oulu 5 Faculty of Biochemistry and Molecular Medicine , , FI-90014 Oulu , Finland
- University of Oulu 6 Faculty of Medicine , , FI-90014 Oulu , Finland
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Research, Center for Biochemistry, Center for Molecular Medicine Cologne (CMMC) 7 , Faculty of Medicine and , Joseph-Stelzmann-Str. 52, 50931 Cologne , Germany
- University Hospital Cologne, University of Cologne 7 , Faculty of Medicine and , Joseph-Stelzmann-Str. 52, 50931 Cologne , Germany
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| |
Collapse
|
20
|
Tucker RP, Degen M. Revisiting the Tenascins: Exploitable as Cancer Targets? Front Oncol 2022; 12:908247. [PMID: 35785162 PMCID: PMC9248440 DOI: 10.3389/fonc.2022.908247] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
For their full manifestation, tumors require support from the surrounding tumor microenvironment (TME), which includes a specific extracellular matrix (ECM), vasculature, and a variety of non-malignant host cells. Together, these components form a tumor-permissive niche that significantly differs from physiological conditions. While the TME helps to promote tumor progression, its special composition also provides potential targets for anti-cancer therapy. Targeting tumor-specific ECM molecules and stromal cells or disrupting aberrant mesenchyme-cancer communications might normalize the TME and improve cancer treatment outcome. The tenascins are a family of large, multifunctional extracellular glycoproteins consisting of four members. Although each have been described to be expressed in the ECM surrounding cancer cells, tenascin-C and tenascin-W are currently the most promising candidates for exploitability and clinical use as they are highly expressed in various tumor stroma with relatively low abundance in healthy tissues. Here, we review what is known about expression of all four tenascin family members in tumors, followed by a more thorough discussion on tenascin-C and tenascin-W focusing on their oncogenic functions and their potential as diagnostic and/or targetable molecules for anti-cancer treatment purposes.
Collapse
Affiliation(s)
- Richard P. Tucker
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA, United States
| | - Martin Degen
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
- *Correspondence: Martin Degen,
| |
Collapse
|
21
|
Fertal SA, Poterala JE, Ponik SM, Wisinski KB. Stromal Characteristics and Impact on New Therapies for Metastatic Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:1238. [PMID: 35267548 PMCID: PMC8909697 DOI: 10.3390/cancers14051238] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/25/2022] Open
Abstract
The heterogenous nature of triple-negative breast cancer (TNBC) is an underlying factor in therapy resistance, metastasis, and overall poor patient outcome. The lack of hormone and growth factor receptors lends to the use of chemotherapy as the first-line treatment for TNBC. However, the failure of chemotherapy demonstrates the need to develop novel immunotherapies, antibody-drug conjugates (ADCs), and other tumor- and stromal-targeted therapeutics for TNBC patients. The potential for stromal-targeted therapy is driven by studies indicating that the interactions between tumor cells and the stromal extracellular matrix (ECM) activate mechanisms of therapy resistance. Here, we will review recent outcomes from clinical trials targeting metastatic TNBC with immunotherapies aimed at programed death ligand-receptor interactions, and ADCs specifically linked to trophoblast cell surface antigen 2 (Trop-2). We will discuss how biophysical and biochemical cues from the ECM regulate the pathophysiology of tumor and stromal cells toward a pro-tumor immune environment, therapy resistance, and poor TNBC patient outcome. Moreover, we will highlight how ECM-mediated resistance is motivating the development of new stromal-targeted therapeutics with potential to improve therapy for this disease.
Collapse
Affiliation(s)
- Shelby A. Fertal
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Johanna E. Poterala
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| | - Suzanne M. Ponik
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Kari B. Wisinski
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| |
Collapse
|
22
|
Yamagata AS, Freire PP, Jones Villarinho N, Teles RHG, Francisco KJM, Jaeger RG, Freitas VM. Transcriptomic Response to Acidosis Reveals Its Contribution to Bone Metastasis in Breast Cancer Cells. Cells 2022; 11:cells11030544. [PMID: 35159353 PMCID: PMC8834614 DOI: 10.3390/cells11030544] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 01/27/2023] Open
Abstract
Bone is the most common site of metastasis in breast cancer. Metastasis is promoted by acidosis, which is associated with osteoporosis. To investigate how acidosis could promote bone metastasis, we compared differentially expressed genes (DEGs) in MDA-MB-231 cancer cells in acidosis, bone metastasis, and bone metastatic tumors. The DEGs were identified using Biojupies and GEO2R. The expression profiles were assessed with Morpheus. The overlapping DEGs between acidosis and bone metastasis were compared to the bulk of the DEGs in terms of the most important genes and enriched terms using CytoHubba and STRING. The expression of the genes in this overlap filtered by secreted proteins was assessed in the osteoporosis secretome. The analysis revealed that acidosis-associated transcriptomic changes were more similar to bone metastasis than bone metastatic tumors. Extracellular matrix (ECM) organization would be the main biological process shared between acidosis and bone metastasis. The secretome genes upregulated in acidosis, bone metastasis, and osteoporosis-associated mesenchymal stem cells are enriched for ECM organization and angiogenesis. Therefore, acidosis may be more important in the metastatic niche than in the primary tumor. Acidosis may contribute to bone metastasis by promoting ECM organization. Untreated osteoporosis could favor bone metastasis through the increased secretion of ECM organization proteins.
Collapse
Affiliation(s)
- Ana Sayuri Yamagata
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (N.J.V.); (R.H.G.T.); (K.J.M.F.); (R.G.J.); (V.M.F.)
- Correspondence:
| | - Paula Paccielli Freire
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil;
| | - Nícolas Jones Villarinho
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (N.J.V.); (R.H.G.T.); (K.J.M.F.); (R.G.J.); (V.M.F.)
| | - Ramon Handerson Gomes Teles
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (N.J.V.); (R.H.G.T.); (K.J.M.F.); (R.G.J.); (V.M.F.)
| | - Kelliton José Mendonça Francisco
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (N.J.V.); (R.H.G.T.); (K.J.M.F.); (R.G.J.); (V.M.F.)
| | - Ruy Gastaldoni Jaeger
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (N.J.V.); (R.H.G.T.); (K.J.M.F.); (R.G.J.); (V.M.F.)
| | - Vanessa Morais Freitas
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (N.J.V.); (R.H.G.T.); (K.J.M.F.); (R.G.J.); (V.M.F.)
| |
Collapse
|
23
|
Loustau T, Abou-Faycal C, Erne W, zur Wiesch PA, Ksouri A, Imhof T, Mörgelin M, Li C, Mathieu M, Salomé N, Crémel G, Dhaouadi S, Bouhaouala-Zahar B, Koch M, Orend G. Modulating tenascin-C functions by targeting the MAtrix REgulating MOtif, “MAREMO”. Matrix Biol 2022; 108:20-38. [DOI: 10.1016/j.matbio.2022.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/31/2022] [Accepted: 02/23/2022] [Indexed: 12/11/2022]
|
24
|
Popova NV, Jücker M. The Functional Role of Extracellular Matrix Proteins in Cancer. Cancers (Basel) 2022; 14:238. [PMID: 35008401 PMCID: PMC8750014 DOI: 10.3390/cancers14010238] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
The extracellular matrix (ECM) is highly dynamic as it is constantly deposited, remodeled and degraded to maintain tissue homeostasis. ECM is a major structural component of the tumor microenvironment, and cancer development and progression require its extensive reorganization. Cancerized ECM is biochemically different in its composition and is stiffer compared to normal ECM. The abnormal ECM affects cancer progression by directly promoting cell proliferation, survival, migration and differentiation. The restructured extracellular matrix and its degradation fragments (matrikines) also modulate the signaling cascades mediated by the interaction with cell-surface receptors, deregulate the stromal cell behavior and lead to emergence of an oncogenic microenvironment. Here, we summarize the current state of understanding how the composition and structure of ECM changes during cancer progression. We also describe the functional role of key proteins, especially tenascin C and fibronectin, and signaling molecules involved in the formation of the tumor microenvironment, as well as the signaling pathways that they activate in cancer cells.
Collapse
Affiliation(s)
- Nadezhda V. Popova
- Laboratory of Receptor Cell Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str., 16/10, 117997 Moscow, Russia;
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| |
Collapse
|
25
|
Sigurdardottir AK, Hilmarsdottir B, Gudjonsson T, Traustadottir GA. Application of 3D Culture Assays to Study Breast Morphogenesis, Epithelial Plasticity, and Cellular Interactions in an Epithelial Progenitor Cell Line. Methods Mol Biol 2022; 2429:391-403. [PMID: 35507176 DOI: 10.1007/978-1-0716-1979-7_26] [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] [Indexed: 06/14/2023]
Abstract
Capturing breast morphogenesis and cancer progression in 3D culture using cell lines with stem cell properties can greatly increase understanding of the underlying mechanisms involved in these processes, highlighting the importance of the culture method. D492 is a breast epithelial progenitor cell line that provides a model for branching morphogenesis when cultured in 3D reconstituted basement membrane matrix (rBM). Along with its derivate cell lines D492M and D492HER2, D492 also serves as a robust model for epithelial to mesenchymal transition (EMT) and tumorigenicity, respectively. Here, we describe the routine maintenance and application of the D492 cell lines in 3D culture for the study of branching morphogenesis, EMT and epithelial-endothelial interaction.
Collapse
Affiliation(s)
- Anna Karen Sigurdardottir
- Stem Cell Research Unit, Department of Anatomy, Faculty of Medicine, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Department of Anatomy, Faculty of Medicine, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Laboratory Hematology, Landspitali-University Hospital, Reykjavik, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Department of Anatomy, Faculty of Medicine, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
26
|
Sun X, Wu B, Chiang HC, Deng H, Zhang X, Xiong W, Liu J, Rozeboom AM, Harris BT, Blommaert E, Gomez A, Garcia RE, Zhou Y, Mitra P, Prevost M, Zhang D, Banik D, Isaacs C, Berry D, Lai C, Chaldekas K, Latham PS, Brantner CA, Popratiloff A, Jin VX, Zhang N, Hu Y, Pujana MA, Curiel TJ, An Z, Li R. Tumour DDR1 promotes collagen fibre alignment to instigate immune exclusion. Nature 2021; 599:673-678. [PMID: 34732895 DOI: 10.1038/s41586-021-04057-2] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 09/22/2021] [Indexed: 12/27/2022]
Abstract
Immune exclusion predicts poor patient outcomes in multiple malignancies, including triple-negative breast cancer (TNBC)1. The extracellular matrix (ECM) contributes to immune exclusion2. However, strategies to reduce ECM abundance are largely ineffective or generate undesired outcomes3,4. Here we show that discoidin domain receptor 1 (DDR1), a collagen receptor with tyrosine kinase activity5, instigates immune exclusion by promoting collagen fibre alignment. Ablation of Ddr1 in tumours promotes the intratumoral penetration of T cells and obliterates tumour growth in mouse models of TNBC. Supporting this finding, in human TNBC the expression of DDR1 negatively correlates with the intratumoral abundance of anti-tumour T cells. The DDR1 extracellular domain (DDR1-ECD), but not its intracellular kinase domain, is required for immune exclusion. Membrane-untethered DDR1-ECD is sufficient to rescue the growth of Ddr1-knockout tumours in immunocompetent hosts. Mechanistically, the binding of DDR1-ECD to collagen enforces aligned collagen fibres and obstructs immune infiltration. ECD-neutralizing antibodies disrupt collagen fibre alignment, mitigate immune exclusion and inhibit tumour growth in immunocompetent hosts. Together, our findings identify a mechanism for immune exclusion and suggest an immunotherapeutic target for increasing immune accessibility through reconfiguration of the tumour ECM.
Collapse
Affiliation(s)
- Xiujie Sun
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Bogang Wu
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Huai-Chin Chiang
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiaowen Zhang
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Junquan Liu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Aaron M Rozeboom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Brent T Harris
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Eline Blommaert
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - Antonio Gomez
- Rheumatology Department and Rheumatology Research Group, Vall d'Hebron Hospital Research Institute, Barcelona, Spain
| | - Roderic Espin Garcia
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - Yufan Zhou
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Payal Mitra
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Madeleine Prevost
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Deyi Zhang
- Department of Medicine, The Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Debarati Banik
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Deborah Berry
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Catherine Lai
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Krysta Chaldekas
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Patricia S Latham
- Department of Pathology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Christine A Brantner
- GW Nanofabrication and Imaging Center, The George Washington University, Washington, DC, USA
| | - Anastas Popratiloff
- GW Nanofabrication and Imaging Center, The George Washington University, Washington, DC, USA
| | - Victor X Jin
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yanfen Hu
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Miguel Angel Pujana
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain.
| | - Tyler J Curiel
- Department of Medicine, The Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA.
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Rong Li
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
27
|
Abstract
INTRODUCTION The matrisome and adhesome comprise proteins that are found within or are associated with the extracellular matrix (ECM) and adhesion complexes, respectively. Interactions between cells and their microenvironment are mediated by key matrisome and adhesome proteins, which direct fundamental processes, including growth and development. Due to their underlying complexity, it has historically been challenging to undertake mass spectrometry (MS)-based profiling of these proteins. New developments in sample preparative workflows, informatics databases, and MS techniques have enabled in-depth proteomic characterization of the matrisome and adhesome, resulting in a comprehensive understanding of the interactomes, and cellular signaling that occur at the cell-ECM interface. AREA COVERED This review summarizes recent advances in proteomic characterization of the matrisome and adhesome. It focuses on the importance of curated databases and discusses key strengths and limitations of different workflows. EXPERT OPINION MS-based proteomics has shown promise in characterizing the matrisome and topology of adhesome networks in health and disease. Moving forward, it will be important to incorporate integrative analysis to define the bidirectional signaling between the matrisome and adhesome, and adopt new methods for post-translational modification and in vivo analyses to better dissect the critical roles that these proteins play in human pathophysiology.
Collapse
Affiliation(s)
- Lukas Krasny
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Paul H Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
28
|
Attaran S, Skoko JJ, Hopkins BL, Wright MK, Wood LE, Asan A, Woo HA, Feinberg A, Neumann CA. Peroxiredoxin-1 Tyr194 phosphorylation regulates LOX-dependent extracellular matrix remodelling in breast cancer. Br J Cancer 2021; 125:1146-1157. [PMID: 34389806 PMCID: PMC8505437 DOI: 10.1038/s41416-021-01510-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/22/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Peroxiredoxin 1 (PRDX1) belongs to an abundant family of peroxidases whose role in cancer is still unresolved. While mouse knockout studies demonstrate a tumour suppressive role for PRDX1, in cancer cell xenografts, results denote PRDX1 as a drug target. Probably, this phenotypic discrepancy stems from distinct roles of PRDX1 in certain cell types or stages of tumour progression. METHODS We demonstrate an important cell-autonomous function for PRDX1 utilising a syngeneic mouse model (BALB/c) and mammary fibroblasts (MFs) obtained from it. RESULTS Loss of PRDX1 in vivo promotes collagen remodelling known to promote breast cancer progression. PRDX1 inactivation in MFs occurs via SRC-induced phosphorylation of PRDX1 TYR194 and not through the expected direct oxidation of CYS52 in PRDX1 by ROS. TYR194-phosphorylated PRDX1 fails to bind to lysyl oxidases (LOX) and leads to the accumulation of extracellular LOX proteins which supports enhanced collagen remodelling associated with breast cancer progression. CONCLUSIONS This study reveals a cell type-specific tumour suppressive role for PRDX1 that is supported by survival analyses, depending on PRDX1 protein levels in breast cancer cohorts.
Collapse
Affiliation(s)
- Shireen Attaran
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - John J Skoko
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Barbara L Hopkins
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Megan K Wright
- University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Laurel E Wood
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alparslan Asan
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Hyun Ae Woo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Adam Feinberg
- Department of Materials Science and Engineering and Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Carola A Neumann
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Trombetta-Lima M, Rosa-Fernandes L, Angeli CB, Moretti IF, Franco YM, Mousessian AS, Wakamatsu A, Lerario AM, Oba-Shinjo SM, Pasqualucci CA, Marie SKN, Palmisano G. Extracellular Matrix Proteome Remodeling in Human Glioblastoma and Medulloblastoma. J Proteome Res 2021; 20:4693-4707. [PMID: 34533964 DOI: 10.1021/acs.jproteome.1c00251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Medulloblastomas (MBs) and glioblastomas (GBMs) are high-incidence central nervous system tumors. Different origin sites and changes in the tissue microenvironment have been associated with the onset and progression. Here, we describe differences between the extracellular matrix (ECM) signatures of these tumors. We compared the proteomic profiles of MB and GBM decellularized tumor samples between each other and their normal decellularized brain site counterparts. Our analysis revealed that 19, 28, and 11 ECM proteins were differentially expressed in MBs, GBMs, and in both MBs and GBMs, respectively. Next, we validated key findings by using a protein tissue array with 53 MB and 55 GBM cases and evaluated the clinical relevance of the identified differentially expressed proteins through their analysis on publicly available datasets, 763 MB samples from the GSE50161 and GSE85217 studies, and 115 GBM samples from RNAseq-TCGA. We report a shift toward a denser fibrillary ECM as well as a clear alteration in the glycoprotein signature, which influences the tumor pathophysiology. MS data have been submitted to the PRIDE repository, project accession: PXD023350.
Collapse
Affiliation(s)
- Marina Trombetta-Lima
- Cellular and Molecular Biology Laboratory (LIM 15), Neurology Department, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Sao Paulo 01246-903, Brazil.,Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen 9713 AV, The Netherlands
| | - Livia Rosa-Fernandes
- Parasitology Department, Instituto de Ciências Biomédicas (ICBUSP), Universidade de Sao Paulo, Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Claudia B Angeli
- Parasitology Department, Instituto de Ciências Biomédicas (ICBUSP), Universidade de Sao Paulo, Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Isabele F Moretti
- Cellular and Molecular Biology Laboratory (LIM 15), Neurology Department, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Sao Paulo 01246-903, Brazil
| | - Yollanda M Franco
- Cellular and Molecular Biology Laboratory (LIM 15), Neurology Department, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Sao Paulo 01246-903, Brazil
| | - Adaliana S Mousessian
- Cellular and Molecular Biology Laboratory (LIM 15), Neurology Department, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Sao Paulo 01246-903, Brazil
| | - Alda Wakamatsu
- Hepatic Pathology Laboratory (LIM 14), Pathology Department, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Sao Paulo 01246-903, Brazil
| | - Antonio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Sueli M Oba-Shinjo
- Cellular and Molecular Biology Laboratory (LIM 15), Neurology Department, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Sao Paulo 01246-903, Brazil
| | - Carlos A Pasqualucci
- Brazilian Aging Brain Study Group, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Sao Paulo 01246-903, Brazil
| | - Suely K N Marie
- Cellular and Molecular Biology Laboratory (LIM 15), Neurology Department, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Sao Paulo 01246-903, Brazil
| | - Giuseppe Palmisano
- Parasitology Department, Instituto de Ciências Biomédicas (ICBUSP), Universidade de Sao Paulo, Sao Paulo, Sao Paulo 05508-000, Brazil
| |
Collapse
|
30
|
Neagu AN, Whitham D, Buonanno E, Jenkins A, Alexa-Stratulat T, Tamba BI, Darie CC. Proteomics and its applications in breast cancer. Am J Cancer Res 2021; 11:4006-4049. [PMID: 34659875 PMCID: PMC8493401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/05/2021] [Indexed: 06/13/2023] Open
Abstract
Breast cancer is an individually unique, multi-faceted and chameleonic disease, an eternal challenge for the new era of high-integrated precision diagnostic and personalized oncomedicine. Besides traditional single-omics fields (such as genomics, epigenomics, transcriptomics and metabolomics) and multi-omics contributions (proteogenomics, proteotranscriptomics or reproductomics), several new "-omics" approaches and exciting proteomics subfields are contributing to basic and advanced understanding of these "multiple diseases termed breast cancer": phenomics/cellomics, connectomics and interactomics, secretomics, matrisomics, exosomics, angiomics, chaperomics and epichaperomics, phosphoproteomics, ubiquitinomics, metalloproteomics, terminomics, degradomics and metadegradomics, adhesomics, stressomics, microbiomics, immunomics, salivaomics, materiomics and other biomics. Throughout the extremely complex neoplastic process, a Breast Cancer Cell Continuum Concept (BCCCC) has been modeled in this review as a spatio-temporal and holistic approach, as long as the breast cancer represents a complex cascade comprising successively integrated populations of heterogeneous tumor and cancer-associated cells, that reflect the carcinoma's progression from a "driving mutation" and formation of the breast primary tumor, toward the distant secondary tumors in different tissues and organs, via circulating tumor cell populations. This BCCCC is widely sustained by a Breast Cancer Proteomic Continuum Concept (BCPCC), where each phenotype of neoplastic and tumor-associated cells is characterized by a changing and adaptive proteomic profile detected in solid and liquid minimal invasive biopsies by complex proteomics approaches. Such a profile is created, beginning with the proteomic landscape of different neoplastic cell populations and cancer-associated cells, followed by subsequent analysis of protein biomarkers involved in epithelial-mesenchymal transition and intravasation, circulating tumor cell proteomics, and, finally, by protein biomarkers that highlight the extravasation and distant metastatic invasion. Proteomics technologies are producing important data in breast cancer diagnostic, prognostic, and predictive biomarkers discovery and validation, are detecting genetic aberrations at the proteome level, describing functional and regulatory pathways and emphasizing specific protein and peptide profiles in human tissues, biological fluids, cell lines and animal models. Also, proteomics can identify different breast cancer subtypes and specific protein and proteoform expression, can assess the efficacy of cancer therapies at cellular and tissular level and can even identify new therapeutic target proteins in clinical studies.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of IașiCarol I bvd. No. 22, Iași 700505, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Emma Buonanno
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Avalon Jenkins
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Teodora Alexa-Stratulat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and PharmacyIndependenței bvd. No. 16-18, Iași 700021, Romania
| | - Bogdan Ionel Tamba
- Advanced Center for Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and PharmacyMihail Kogălniceanu Street No. 9-13, Iași 700454, Romania
| | - Costel C Darie
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| |
Collapse
|
31
|
Schuler LA, Murdoch FE. Endogenous and Therapeutic Estrogens: Maestro Conductors of the Microenvironment of ER+ Breast Cancers. Cancers (Basel) 2021; 13:3725. [PMID: 34359625 PMCID: PMC8345134 DOI: 10.3390/cancers13153725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/25/2022] Open
Abstract
Estrogen receptor alpha (ERα) marks heterogeneous breast cancers which display a repertoire of somatic genomic mutations and an immune environment that differs from other breast cancer subtypes. These cancers also exhibit distinct biological behaviors; despite an overall better prognosis than HER2+ or triple negative breast cancers, disseminated dormant cells can lead to disease recurrence decades after the initial diagnosis and treatment. Estrogen is the best studied driver of these cancers, and antagonism or reduction of estrogen activity is the cornerstone of therapeutic approaches. In addition to reducing proliferation of ERα+ cancer cells, these treatments also alter signals to multiple other target cells in the environment, including immune cell subpopulations, cancer-associated fibroblasts, and endothelial cells via several distinct estrogen receptors. In this review, we update progress in our understanding of the stromal cells populating the microenvironments of primary and metastatic ER+ tumors, the effects of estrogen on tumor and stromal cells to modulate immune activity and the extracellular matrix, and net outcomes in experimental and clinical studies. We highlight new approaches that will illuminate the unique biology of these cancers, provide the foundation for developing new treatment and prevention strategies, and reduce mortality of this disease.
Collapse
Affiliation(s)
- Linda A. Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | | |
Collapse
|
32
|
Analysis of Colorectal Carcinogenesis Paradigm between Cold Constitution and Heat Constitution: Earlier ECM Collagen Deposition. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5547578. [PMID: 34335820 PMCID: PMC8313331 DOI: 10.1155/2021/5547578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/26/2021] [Accepted: 07/09/2021] [Indexed: 11/17/2022]
Abstract
Colorectal cancer (CRC) is a common malignant tumor around the world. Studying the unique constitution of CRC patients is conducive to the application of personalized medical treatment for CRC. The most common types of constitution in CRC are cold and heat constitution. A previous study has suggested that the malignant progression in cold and heat constitution CRC are different; however, the mechanism remains unclear. The tumor microenvironment (TME) is likely to vary with each individual constitution, which may affect the tumor growth in different constitutions. The extracellular matrix (ECM), the most important component of TME, plays a critical role in disease progression and outcome in patients with CRC. Moreover, collagen, the major component of the ECM, determines the main functional characteristics of ECM and tissue fibrosis caused by collagen deposition, which is one of the signs of CRC malignant progression. This study aimed to explore the mechanisms leading to different colorectal carcinogenesis paradigms between the cold constitution and heat constitution within the context of ECM collagen deposition. We established the CRC rat models and enrolled 30 CRC patients with cold and heat constitution. The collagen-related parameters were detected by using Sirius red staining combined with polarized light microscope, and expressions of collagen (COL I and COL III) and lysyl oxidase (LOX and LOXL2) were determined using immunohistochemistry, while the mRNA levels of COL1A1, COL3A1, LOX, and LOXL2 were measured by qRT-PCR. We found that a higher degree of collagen deposition in the cold-constitution group. The results suggest cold and heat constitution may affect the colorectal carcinogenesis paradigm by influencing the early collagen deposition in colon tissue. The study may provide an effective idea for clinicians to improve the prognosis of CRC patients with different constitutions.
Collapse
|
33
|
Directional cues in the tumor microenvironment due to cell contraction against aligned collagen fibers. Acta Biomater 2021; 129:96-109. [PMID: 33965625 PMCID: PMC8848478 DOI: 10.1016/j.actbio.2021.04.053] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
It is well established that collagen alignment in the breast tumor microenvironment provides biophysical cues to drive disease progression. Numerous mechanistic studies have demonstrated that tumor cell behavior is driven by the architecture and stiffness of the collagen matrix. However, the mechanical properties within a 3D collagen microenvironment, particularly at the scale of the cell, remain poorly defined. To investigate cell-scale mechanical cues with respect to local collagen architecture, we employed a combination of intravital imaging of the mammary tumor microenvironment and a 3D collagen gel system with both acellular pNIPAAm microspheres and MDA-MB-231 breast carcinoma cells. Within the in vivo tumor microenvironment, the displacement of collagen fiber was identified in response to tumor cells migrating through the stromal matrix. To further investigate cell-scale stiffness in aligned fiber architectures and the propagation of cell-induced fiber deformations, precise control of collagen architecture was coupled with innovative methodology to measure mechanical properties of the collagen fiber network. This method revealed up to a 35-fold difference in directional cell-scale stiffness resulting from contraction against aligned fibers. Furthermore, the local anisotropy of the matrix dramatically altered the rate at which contractility-induced fiber displacements decayed over distance. Together, our results reveal mechanical properties in aligned matrices that provide dramatically different cues to the cell in perpendicular directions. These findings are supported by the mechanosensing behavior of tumor cells and have important implications for cell-cell communication within the tissue microenvironment. STATEMENT OF SIGNIFICANCE: It is widely appreciated that the architecture of the extracellular matrix impacts cellular behavior in normal and disease states. Numerous studies have determined the fundamental role of collagen matrix architecture on cellular mechanosensing, but effectively quantifying anisotropic mechanical properties of the collagen matrix at the cell-scale remains challenging. Here, we developed innovative methodology to discover that collagen alignment results in a 35-fold difference in cell-scale stiffness and alters contractile force transmission through the fiber network. Furthermore, we identified bias in cell response along the axis of alignment, where local stiffness is highest. Overall, our results define cell-scale stiffness and fiber deformations due to collagen architecture that may instruct cell communication within a broad range of tissue microenvironments.
Collapse
|
34
|
Murdamoothoo D, Sun Z, Yilmaz A, Riegel G, Abou‐Faycal C, Deligne C, Velazquez‐Quesada I, Erne W, Nascimento M, Mörgelin M, Cremel G, Paul N, Carapito R, Veber R, Dumortier H, Yuan J, Midwood KS, Loustau T, Orend G. Tenascin-C immobilizes infiltrating T lymphocytes through CXCL12 promoting breast cancer progression. EMBO Mol Med 2021; 13:e13270. [PMID: 33988305 PMCID: PMC8185552 DOI: 10.15252/emmm.202013270] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint therapy, where CD8 tumor infiltrating T lymphocytes (TIL) are reactivated, is a promising anti-cancer treatment approach, yet with low response rates. The extracellular matrix, in particular tenascin-C, may generate barriers for TIL. To investigate this possibility, we used a MMTV-NeuNT and syngeneic mammary gland grafting model derived thereof with engineered tenascin-C levels and observed accumulation of CD8 TIL in tenascin-C-rich stroma. Inhibition studies revealed that tenascin-C induced CXCL12 through TLR4. By binding CXCL12, tenascin-C retained CD8 TIL in the stroma. Blockade of CXCR4, the receptor of CXCL12, enhanced macrophage and CD8 TIL infiltration and reduced tumor growth and subsequent metastasis. Retention of CD8 TIL by tenascin-C/CXCL12 was also observed in human breast cancer by tissue staining. Moreover, whereas high CD8 TIL numbers correlated with longer metastasis-free survival, this was not the case when also tenascin-C and CXCL12 levels were high. Altogether, these results may be useful for improving tumor immunity as diagnostic tool and to formulate a future "TIL-matrix-release-and-reactivate" strategy.
Collapse
|
35
|
Abstract
The extracellular matrix is a fundamental, core component of all tissues and organs, and is essential for the existence of multicellular organisms. From the earliest stages of organism development until death, it regulates and fine-tunes every cellular process in the body. In cancer, the extracellular matrix is altered at the biochemical, biomechanical, architectural and topographical levels, and recent years have seen an exponential increase in the study and recognition of the importance of the matrix in solid tumours. Coupled with the advancement of new technologies to study various elements of the matrix and cell-matrix interactions, we are also beginning to see the deployment of matrix-centric, stromal targeting cancer therapies. This Review touches on many of the facets of matrix biology in solid cancers, including breast, pancreatic and lung cancer, with the aim of highlighting some of the emerging interactions of the matrix and influences that the matrix has on tumour onset, progression and metastatic dissemination, before summarizing the ongoing work in the field aimed at developing therapies to co-target the matrix in cancer and cancer metastasis.
Collapse
Affiliation(s)
- Thomas R Cox
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
36
|
Holstein E, Dittmann A, Kääriäinen A, Pesola V, Koivunen J, Pihlajaniemi T, Naba A, Izzi V. The Burden of Post-Translational Modification (PTM)-Disrupting Mutations in the Tumor Matrisome. Cancers (Basel) 2021; 13:1081. [PMID: 33802493 PMCID: PMC7959462 DOI: 10.3390/cancers13051081] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND To evaluate the occurrence of mutations affecting post-translational modification (PTM) sites in matrisome genes across different tumor types, in light of their genomic and functional contexts and in comparison with the rest of the genome. METHODS This study spans 9075 tumor samples and 32 tumor types from The Cancer Genome Atlas (TCGA) Pan-Cancer cohort and identifies 151,088 non-silent mutations in the coding regions of the matrisome, of which 1811 affecting known sites of hydroxylation, phosphorylation, N- and O-glycosylation, acetylation, ubiquitylation, sumoylation and methylation PTM. RESULTS PTM-disruptive mutations (PTMmut) in the matrisome are less frequent than in the rest of the genome, seem independent of cell-of-origin patterns but show dependence on the nature of the matrisome protein affected and the background PTM types it generally harbors. Also, matrisome PTMmut are often found among structural and functional protein regions and in proteins involved in homo- and heterotypic interactions, suggesting potential disruption of matrisome functions. CONCLUSIONS Though quantitatively minoritarian in the spectrum of matrisome mutations, PTMmut show distinctive features and damaging potential which might concur to deregulated structural, functional, and signaling networks in the tumor microenvironment.
Collapse
Affiliation(s)
- Elisa Holstein
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Annalena Dittmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Anni Kääriäinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Vilma Pesola
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Jarkko Koivunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Taina Pihlajaniemi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA;
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
- Faculty of Medicine, University of Oulu, FI-90014 Oulu, Finland
- Finnish Cancer Institute, 00130 Helsinki, Finland
| |
Collapse
|
37
|
Ouellette JN, Drifka CR, Pointer KB, Liu Y, Lieberthal TJ, Kao WJ, Kuo JS, Loeffler AG, Eliceiri KW. Navigating the Collagen Jungle: The Biomedical Potential of Fiber Organization in Cancer. Bioengineering (Basel) 2021; 8:17. [PMID: 33494220 PMCID: PMC7909776 DOI: 10.3390/bioengineering8020017] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Recent research has highlighted the importance of key tumor microenvironment features, notably the collagen-rich extracellular matrix (ECM) in characterizing tumor invasion and progression. This led to great interest from both basic researchers and clinicians, including pathologists, to include collagen fiber evaluation as part of the investigation of cancer development and progression. Fibrillar collagen is the most abundant in the normal extracellular matrix, and was revealed to be upregulated in many cancers. Recent studies suggested an emerging theme across multiple cancer types in which specific collagen fiber organization patterns differ between benign and malignant tissue and also appear to be associated with disease stage, prognosis, treatment response, and other clinical features. There is great potential for developing image-based collagen fiber biomarkers for clinical applications, but its adoption in standard clinical practice is dependent on further translational and clinical evaluations. Here, we offer a comprehensive review of the current literature of fibrillar collagen structure and organization as a candidate cancer biomarker, and new perspectives on the challenges and next steps for researchers and clinicians seeking to exploit this information in biomedical research and clinical workflows.
Collapse
Affiliation(s)
- Jonathan N. Ouellette
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
| | - Cole R. Drifka
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
| | - Kelli B. Pointer
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
| | - Yuming Liu
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
| | - Tyler J Lieberthal
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
| | - W John Kao
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
- Department of Industrial and Manufacturing Systems Engineering, Faculty of Engineering, University of Hong Kong, Pokfulam, Hong Kong
| | - John S. Kuo
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Agnes G. Loeffler
- Department of Pathology, MetroHealth Medical Center, Cleveland, OH 44109, USA;
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
| |
Collapse
|
38
|
Knott SJ, Brown KA, Josyer H, Carr A, Inman D, Jin S, Friedl A, Ponik SM, Ge Y. Photocleavable Surfactant-Enabled Extracellular Matrix Proteomics. Anal Chem 2020; 92:15693-15698. [PMID: 33232116 DOI: 10.1021/acs.analchem.0c03104] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) provides an architectural meshwork that surrounds and supports cells. The dysregulation of heavily post-translationally modified ECM proteins directly contributes to various diseases. Mass spectrometry (MS)-based proteomics is an ideal tool to identify ECM proteins and characterize their post-translational modifications, but ECM proteomics remains challenging owing to the extremely low solubility of the ECM. Herein, enabled by effective solubilization of ECM proteins using our recently developed photocleavable surfactant, Azo, we have developed a streamlined ECM proteomic strategy that allows fast tissue decellularization, efficient extraction and enrichment of ECM proteins, and rapid digestion prior to reversed-phase liquid chromatography (RPLC)-MS analysis. A total of 173 and 225 unique ECM proteins from mouse mammary tumors have been identified using 1D and 2D RPLC-MS/MS, respectively. Moreover, 87 (from 1DLC-MS/MS) and 229 (from 2DLC-MS/MS) post-translational modifications of ECM proteins, including glycosylation, phosphorylation, and hydroxylation, were identified and localized. This Azo-enabled ECM proteomics strategy will streamline the analysis of ECM proteins and promote the study of ECM biology.
Collapse
Affiliation(s)
- Samantha J Knott
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Kyle A Brown
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Harini Josyer
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Austin Carr
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - David Inman
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Song Jin
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Andreas Friedl
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1685 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States.,Human Proteomics Program, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| |
Collapse
|
39
|
Kääriäinen A, Pesola V, Dittmann A, Kontio J, Koivunen J, Pihlajaniemi T, Izzi V. Machine Learning Identifies Robust Matrisome Markers and Regulatory Mechanisms in Cancer. Int J Mol Sci 2020; 21:E8837. [PMID: 33266472 PMCID: PMC7700160 DOI: 10.3390/ijms21228837] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
The expression and regulation of matrisome genes-the ensemble of extracellular matrix, ECM, ECM-associated proteins and regulators as well as cytokines, chemokines and growth factors-is of paramount importance for many biological processes and signals within the tumor microenvironment. The availability of large and diverse multi-omics data enables mapping and understanding of the regulatory circuitry governing the tumor matrisome to an unprecedented level, though such a volume of information requires robust approaches to data analysis and integration. In this study, we show that combining Pan-Cancer expression data from The Cancer Genome Atlas (TCGA) with genomics, epigenomics and microenvironmental features from TCGA and other sources enables the identification of "landmark" matrisome genes and machine learning-based reconstruction of their regulatory networks in 74 clinical and molecular subtypes of human cancers and approx. 6700 patients. These results, enriched for prognostic genes and cross-validated markers at the protein level, unravel the role of genetic and epigenetic programs in governing the tumor matrisome and allow the prioritization of tumor-specific matrisome genes (and their regulators) for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Anni Kääriäinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Vilma Pesola
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Annalena Dittmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Juho Kontio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Jarkko Koivunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Taina Pihlajaniemi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
- Faculty of Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland
- Finnish Cancer Institute, 00130 Helsinki, Finland
| |
Collapse
|
40
|
Micek HM, Visetsouk MR, Masters KS, Kreeger PK. Engineering the Extracellular Matrix to Model the Evolving Tumor Microenvironment. iScience 2020; 23:101742. [PMID: 33225247 PMCID: PMC7666341 DOI: 10.1016/j.isci.2020.101742] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinical evidence supports a role for the extracellular matrix (ECM) in cancer risk and prognosis across multiple tumor types, and numerous studies have demonstrated that individual ECM components impact key hallmarks of tumor progression (e.g., proliferation, migration, angiogenesis). However, the ECM is a complex network of fibrillar proteins, glycoproteins, and proteoglycans that undergoes dramatic changes in composition and organization during tumor development. In this review, we will highlight how engineering approaches can be used to examine the impact of changes in tissue architecture, ECM composition (i.e., identity and levels of individual ECM components), and cellular- and tissue-level mechanics on tumor progression. In addition, we will discuss recently developed methods to model the ECM that have not yet been applied to the study of cancer.
Collapse
Affiliation(s)
- Hannah M. Micek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mike R. Visetsouk
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kristyn S. Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Pamela K. Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
41
|
Izzi V, Davis MN, Naba A. Pan-Cancer Analysis of the Genomic Alterations and Mutations of the Matrisome. Cancers (Basel) 2020; 12:E2046. [PMID: 32722287 PMCID: PMC7463652 DOI: 10.3390/cancers12082046] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/17/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM) is a master regulator of all cellular functions and a major component of the tumor microenvironment. We previously defined the "matrisome" as the ensemble of genes encoding ECM proteins and proteins modulating ECM structure or function. While compositional and biomechanical changes in the ECM regulate cancer progression, no study has investigated the genomic alterations of matrisome genes in cancers and their consequences. Here, mining The Cancer Genome Atlas (TCGA) data, we found that copy number alterations and mutations are frequent in matrisome genes, even more so than in the rest of the genome. We also found that these alterations are predicted to significantly impact gene expression and protein function. Moreover, we identified matrisome genes whose mutational burden is an independent predictor of survival. We propose that studying genomic alterations of matrisome genes will further our understanding of the roles of this compartment in cancer progression and will lead to the development of innovative therapeutic strategies targeting the ECM.
Collapse
Affiliation(s)
- Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland;
- Finnish Cancer Institute, 00130 Helsinki, Finland
| | - Martin N. Davis
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA;
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
42
|
Brett EA, Sauter MA, Machens HG, Duscher D. Tumor-associated collagen signatures: pushing tumor boundaries. Cancer Metab 2020; 8:14. [PMID: 32637098 PMCID: PMC7331261 DOI: 10.1186/s40170-020-00221-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/23/2020] [Indexed: 01/19/2023] Open
Abstract
In 2006, a new model of invasive breast tumor emerged and, since 2011, is gaining recognition and research momentum. "Tumor-associated collagen signatures" describe 3 distinct layers of collagen which radiate outward in shells from the main body of the tumor. The outermost layer (TACS3) features branches of collagen radiating away from the tumor, 90° perpendicular to the tumor surface. TACS3 increases tumor span and correlates directly with metastasis, though presently difficult to detect in breast tissue. TACS is an emerging model but has been validated by multiple labs in vitro and in vivo, specifically for breast cancer prognostics. Newly recognized and accepted tumor borders will impact both R0 resections and downstream surgical reconstruction. This review aims to comprehensively introduce and connect the ranging literature on linearized collagen of invasive tumor borders. Using PubMed keyword searches containing "aligned," "linear," "oriented," and "organized," we have gathered the studies on TACS, integrated the concept into the clinic, and projected future platforms.
Collapse
Affiliation(s)
- Elizabeth A Brett
- Department of Plastic and Hand Surgery, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Matthias A Sauter
- Department of Plastic and Hand Surgery, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Dominik Duscher
- Department of Plastic and Hand Surgery, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| |
Collapse
|
43
|
Naranjo JD, Saldin LT, Sobieski E, Quijano LM, Hill RC, Chan PG, Torres C, Dziki JL, Cramer MC, Lee YC, Das R, Bajwa AK, Nossair R, Klimak M, Marchal L, Patel S, Velankar SS, Hansen KC, McGrath K, Badylak SF. Esophageal extracellular matrix hydrogel mitigates metaplastic change in a dog model of Barrett's esophagus. SCIENCE ADVANCES 2020; 6:eaba4526. [PMID: 32656339 PMCID: PMC7329334 DOI: 10.1126/sciadv.aba4526] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/16/2020] [Indexed: 05/17/2023]
Abstract
Chronic inflammatory gastric reflux alters the esophageal microenvironment and induces metaplastic transformation of the epithelium, a precancerous condition termed Barrett's esophagus (BE). The microenvironmental niche, which includes the extracellular matrix (ECM), substantially influences cell phenotype. ECM harvested from normal porcine esophageal mucosa (eECM) was formulated as a mucoadhesive hydrogel, and shown to largely retain basement membrane and matrix-cell adhesion proteins. Dogs with BE were treated orally with eECM hydrogel and omeprazole (n = 6) or omeprazole alone (n = 2) for 30 days. eECM treatment resolved esophagitis, reverted metaplasia to a normal, squamous epithelium in four of six animals, and downregulated the pro-inflammatory tumor necrosis factor-α+ cell infiltrate compared to control animals. The metaplastic tissue in control animals (n = 2) did not regress. The results suggest that in vivo alteration of the microenvironment with a site-appropriate, mucoadhesive ECM hydrogel can mitigate the inflammatory and metaplastic response in a dog model of BE.
Collapse
Affiliation(s)
- Juan Diego Naranjo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lindsey T. Saldin
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Eric Sobieski
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Lina M. Quijano
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Ryan C. Hill
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Patrick G. Chan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Cardiothoracic Surgery, UPMC, Pittsburgh, PA 15213, USA
| | - Crisanto Torres
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Cardiothoracic Surgery, UPMC, Pittsburgh, PA 15213, USA
| | - Jenna L. Dziki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Madeline C. Cramer
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yoojin C. Lee
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rohit Das
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, UPMC, Pittsburgh, PA 15213, USA
| | - Anant K. Bajwa
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Rania Nossair
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Molly Klimak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Lucile Marchal
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Shil Patel
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Sachin S. Velankar
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical Engineering, Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kevin McGrath
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, UPMC, Pittsburgh, PA 15213, USA
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
44
|
The significance of stromal collagen organization in cancer tissue: An in-depth discussion of literature. Crit Rev Oncol Hematol 2020; 151:102907. [DOI: 10.1016/j.critrevonc.2020.102907] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/09/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
|
45
|
Lugo-Cintrón KM, Ayuso JM, White BR, Harari PM, Ponik S, Beebe DJ, Gong MM, Virumbrales-Muñoz M. Matrix density drives 3D organotypic lymphatic vessel activation in a microfluidic model of the breast tumor microenvironment. LAB ON A CHIP 2020; 20:1586-1600. [PMID: 32297896 PMCID: PMC7330815 DOI: 10.1039/d0lc00099j] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Lymphatic vessels (LVs) have been suggested as a preferential conduit for metastatic progression in breast cancer, where a correlation between the occurrence of lymph node metastasis and an increased extracellular matrix (ECM) density has been reported. However, the effect of ECM density on LV function is largely unknown. To better understand these effects, we used a microfluidic device to recreate tubular LVs in a collagen type I matrix. The density of the matrix was tailored to mimic normal breast tissue using a low-density collagen (LD-3 mg mL-1) and cancerous breast tissue using a high-density collagen (HD-6 mg mL-1). We investigated the effect of ECM density on LV morphology, growth, cytokine secretion, and barrier function. LVs cultured in HD matrices showed morphological changes as compared to LVs cultured in a LD matrix. Specifically, LVs cultured in HD matrices had a 3-fold higher secretion of the pro-inflammatory cytokine, IL-6, and a leakier phenotype, suggesting LVs acquired characteristics of activated vessels. Interestingly, LV leakiness was mitigated by blocking the IL-6 receptor on the lymphatic ECs, maintaining endothelium permeability at similar levels of LV cultured in a LD matrix. To recreate a more in vivo microenvironment, we incorporated metastatic breast cancer cells (MDA-MB-231) into the LD and HD matrices. For HD matrices, co-culture with MDA-MB-231 cells exacerbated vessel leakiness and secretion of IL-6. In summary, our data suggest that (1) ECM density is an important microenvironmental cue that affects LV function in the breast tumor microenvironment (TME), (2) dense matrices condition LVs towards an activated phenotype and (3) blockade of IL-6 signaling may be a potential therapeutic target to mitigate LV dysfunction. Overall, modeling LVs and their interactions with the TME can help identify novel therapeutic targets and, in turn, advance therapeutic discovery.
Collapse
Affiliation(s)
- Karina M. Lugo-Cintrón
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - José M. Ayuso
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Bridget R. White
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Suzanne Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - David J. Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Max M. Gong
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - María Virumbrales-Muñoz
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
46
|
Breast Fibroblasts and ECM Components Modulate Breast Cancer Cell Migration Through the Secretion of MMPs in a 3D Microfluidic Co-Culture Model. Cancers (Basel) 2020; 12:cancers12051173. [PMID: 32384738 PMCID: PMC7281408 DOI: 10.3390/cancers12051173] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/28/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023] Open
Abstract
The extracellular matrix (ECM) composition greatly influences cancer progression, leading to differential invasion, migration, and metastatic potential. In breast cancer, ECM components, such as fibroblasts and ECM proteins, have the potential to alter cancer cell migration. However, the lack of in vitro migration models that can vary ECM composition limits our knowledge of how specific ECM components contribute to cancer progression. Here, a microfluidic model was used to study the effect of 3D heterogeneous ECMs (i.e., fibroblasts and different ECM protein compositions) on the migration distance of a highly invasive human breast cancer cell line, MDA-MB-231. Specifically, we show that in the presence of normal breast fibroblasts, a fibronectin-rich matrix induces more cancer cell migration. Analysis of the ECM revealed the presence of ECM tunnels. Likewise, cancer-stromal crosstalk induced an increase in the secretion of metalloproteinases (MMPs) in co-cultures. When MMPs were inhibited, migration distance decreased in all conditions except for the fibronectin-rich matrix in the co-culture with human mammary fibroblasts (HMFs). This model mimics the in vivo invasion microenvironment, allowing the examination of cancer cell migration in a relevant context. In general, this data demonstrates the capability of the model to pinpoint the contribution of different components of the tumor microenvironment (TME).
Collapse
|
47
|
Frid MG, Thurman JM, Hansen KC, Maron BA, Stenmark KR. Inflammation, immunity, and vascular remodeling in pulmonary hypertension; Evidence for complement involvement? Glob Cardiol Sci Pract 2020; 2020:e202001. [PMID: 32478115 PMCID: PMC7232865 DOI: 10.21542/gcsp.2020.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary (arterial) hypertension (PH/PAH) is a life-threatening cardiopulmonary disorder. Experimental evidence suggests involvement of inflammatory and autoimmune processes in pathogenesis of PH/PAH, however the triggering and disease-promoting mechanisms remain unknown. The complement system is a key arm of innate immunity implicated in various pro-inflammatory and autoimmune diseases, yet, surprisingly little is known about the role of complement in PH/PAH pathogenesis. The preponderance of the existing data associates complement with PH/PAH via analysis of plasma and does not study the lung directly. Therefore, we aimed to resolve this by analyzing both the mechanisms of local lung-specific complement activation and the correlation of dysregulated plasma complement to clinical outcome in PAH patients. In our recent studies, reviewed herein, we show, for the first time, that immunoglobulin-driven activation of the complement cascade, specifically its alternative pathway, in the pulmonary perivascular areas, is a key mechanism initiating pro-inflammatory processes in the early stage of experimental hypoxic PH (a form of "sterile inflammation"). In human patients with end-stage PAH, we have demonstrated that perivascular deposition of immunoglobulin G (IgG) and activation of the complement cascade are "longitudinally" persistent in the disease. We also showed, using unbiased network analysis, that plasma complement signaling, including again the Alternative pathway, is a prognostic factor of survival in patients with idiopathic PAH (IPAH). Based on these initial findings, we suggest that vascular-specific, immunoglobulin-driven dysregulated complement signaling triggers and maintains pulmonary vascular remodeling and PH. Future experiments in this area would facilitate discoveries on whether complement signaling can serve both as a biomarker and therapeutic target in PH/PAH.
Collapse
Affiliation(s)
- Maria G. Frid
- University of Colorado, Denver, Anschutz Medical Campus, USA
| | | | - Kirk C. Hansen
- University of Colorado, Denver, Anschutz Medical Campus, USA
| | | | | |
Collapse
|
48
|
Gerarduzzi C, Hartmann U, Leask A, Drobetsky E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res 2020; 80:2705-2717. [PMID: 32193287 DOI: 10.1158/0008-5472.can-18-2098] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2019] [Accepted: 03/17/2020] [Indexed: 11/16/2022]
Abstract
The extracellular matrix (ECM) surrounding cells is indispensable for regulating their behavior. The dynamics of ECM signaling are tightly controlled throughout growth and development. During tissue remodeling, matricellular proteins (MCP) are secreted into the ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins and cell-matrix interactions to influence normal cellular functions. In the tumor microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can support multiple hallmarks of carcinogenesis by interacting with various cellular components that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This realization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer, as well as effective and selective therapeutic targets.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada. .,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Elliot Drobetsky
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
49
|
Brechbuhl HM, Barrett AS, Kopin E, Hagen JC, Han AL, Gillen AE, Finlay-Schultz J, Cittelly DM, Owens P, Horwitz KB, Sartorius CA, Hansen K, Kabos P. Fibroblast subtypes define a metastatic matrisome in breast cancer. JCI Insight 2020; 5:130751. [PMID: 32045383 DOI: 10.1172/jci.insight.130751] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 01/29/2020] [Indexed: 02/06/2023] Open
Abstract
Small primary breast cancers can show surprisingly high potential for metastasis. Clinical decision-making for tumor aggressiveness, including molecular profiling, relies primarily on analysis of the cancer cells. Here we show that this analysis is insufficient - that the stromal microenvironment of the primary tumor plays a key role in tumor cell dissemination and implantation at distant sites. We previously described 2 cancer-associated fibroblasts (CAFs) that either express (CD146+) or lack (CD146-) CD146 (official symbol MCAM, alias MUC18). We now find that when mixed with human breast cancer cells, each fibroblast subtype determines the fate of cancer cells: CD146- fibroblasts promoted increased metastasis compared with CD146+ fibroblasts. Potentially novel quantitative and qualitative proteomic analyses showed that CD146+ CAFs produced an environment rich in basement membrane proteins, while CD146- CAFs exhibited increases in fibronectin 1, lysyl oxidase, and tenascin C, all overexpressed in aggressive disease. We also show clinically that CD146- CAFs predicted for likelihood of lymph node involvement even in small primary tumors (<5 cm). Clearly small tumors enriched for CD146- CAFs require aggressive treatments.
Collapse
Affiliation(s)
| | | | - Etana Kopin
- Division of Medical Oncology, Department of Medicine
| | - Jaime C Hagen
- Division of Medical Oncology, Department of Medicine
| | - Amy L Han
- Division of Medical Oncology, Department of Medicine
| | | | - Jessica Finlay-Schultz
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado USA
| | - Diana M Cittelly
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado USA
| | - Philip Owens
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado USA.,Research Service, Department of Veterans Affairs, Eastern Colorado Health Care System, Aurora, Colorado, USA
| | - Kathryn B Horwitz
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado USA.,Division of Endocrinology, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics
| | - Peter Kabos
- Division of Medical Oncology, Department of Medicine
| |
Collapse
|
50
|
Izzi V, Koivunen J, Rappu P, Heino J, Pihlajaniemi T. Integration of Matrisome Omics: Towards System Biology of the Tumor Matrisome. EXTRACELLULAR MATRIX OMICS 2020. [DOI: 10.1007/978-3-030-58330-9_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|