1
|
Tang X, Liu Y, Zhao J, Fu C, Yang W. Subtyping of gastric cancer based on basement membrane genes that stratifies the prognosis, immune infiltration and therapeutic response. Discov Oncol 2024; 15:362. [PMID: 39164593 PMCID: PMC11336019 DOI: 10.1007/s12672-024-01238-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
Gastric cancer (GC) is highly heterogeneous and prone to metastasis, which are obstacles to the effectiveness of treatment. The basement membrane (BM) acts as a barrier to tumor cell invasion and metastasis. It is critical to investigate the relationship between BM status, metastasis, and patient prognosis. In several large cohorts, we investigated BM gene expression-based molecular classification and risk-prognosis models for GC, examined tumor microenvironment (TME) differences among different molecular subtypes, and developed risk models in predicting prognosis, immunotherapy effectiveness, and chemotherapy resistance. Three GC subtypes (BMclusterA/B/C) based on BM gene expression status were discovered. Each of the three GC subtypes has unique immune infiltration and activated oncogenic signals. Moreover, a 6-gene score (BMscore) predictive model was developed. The low BMscore group had a high tumor mutation burden, high immunogenicity, and low RHOJ expression levels, implying that individuals with GC in this category may be more susceptible to immunotherapy and treatment. The EMT subtype showed a considerably higher BMscore than the other subtypes in the Asian Organization for Research on Cancer (ACRG) molecular classification. Endothelial cells, smooth muscle cells, and fibroblasts may be engaged in regulating BM reorganization in GC progression, according to single-cell transcriptome analyses. In conclusion, we defined a novel molecular classification of GC based on BM genes, developed a prognostic risk model, and elucidated the cell subpopulations involved in BM remodeling at the single-cell level. This study has deepened the understanding of the relationship between GC metastasis and BM alterations, achieved prognostic stratification, and guided therapy.
Collapse
Affiliation(s)
- Xin Tang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushanhu Road, Hefei, 230031, Anhui, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230026, China
| | - Yu Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushanhu Road, Hefei, 230031, Anhui, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230026, China
| | - Jiarong Zhao
- Medical Pathology Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Changfang Fu
- Department of Pharmacy, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China.
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, 230001, Anhui, China.
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushanhu Road, Hefei, 230031, Anhui, China.
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
2
|
Lim SE, Vicente-Munuera P, Mao Y. Forced back into shape: Mechanics of epithelial wound repair. Curr Opin Cell Biol 2024; 87:102324. [PMID: 38290420 DOI: 10.1016/j.ceb.2024.102324] [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: 10/06/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
Wound repair, the closing of a hole, is inherently a physical process that requires the change of shape of materials, in this case, cells and tissues. Not only is efficient and accurate wound repair critical for restoring barrier function and reducing infection, but it is also critical for restoring the complex three-dimensional architecture of an organ. This re-sculpting of tissues requires the complex coordination of cell behaviours in multiple dimensions, in space and time, to ensure that the repaired structure can continue functioning optimally. Recent evidence highlights the importance of cell and tissue mechanics in 2D and 3D to achieve such seamless wound repair.
Collapse
Affiliation(s)
- Shu En Lim
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK
| | - Pablo Vicente-Munuera
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK
| | - Yanlan Mao
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
3
|
Bedelbaeva K, Cameron B, Latella J, Aslanukov A, Gourevitch D, Davuluri R, Heber-Katz E. Epithelial-mesenchymal transition: an organizing principle of mammalian regeneration. Front Cell Dev Biol 2023; 11:1101480. [PMID: 37965571 PMCID: PMC10641390 DOI: 10.3389/fcell.2023.1101480] [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: 11/17/2022] [Accepted: 09/27/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction: The MRL mouse strain is one of the few examples of a mammal capable of healing appendage wounds by regeneration, a process that begins with the formation of a blastema, a structure containing de-differentiating mesenchymal cells. HIF-1α expression in the nascent MRL wound site blastema is one of the earliest identified events and is sufficient to initiate the complete regenerative program. However, HIF-1α regulates many cellular processes modulating the expression of hundreds of genes. A later signal event is the absence of a functional G1 checkpoint, leading to G2 cell cycle arrest with increased cellular DNA but little cell division observed in the blastema. This lack of mitosis in MRL blastema cells is also a hallmark of regeneration in classical invertebrate and vertebrate regenerators such as planaria, hydra, and newt. Results and discussion: Here, we explore the cellular events occurring between HIF-1α upregulation and its regulation of the genes involved in G2 arrest (EVI-5, γH3, Wnt5a, and ROR2), and identify epithelial-mesenchymal transition (EMT) (Twist and Slug) and chromatin remodeling (EZH-2 and H3K27me3) as key intermediary processes. The locus of these cellular events is highly regionalized within the blastema, occurring in the same cells as determined by double staining by immunohistochemistry and FACS analysis, and appears as EMT and chromatin remodeling, followed by G2 arrest determined by kinetic expression studies.
Collapse
Affiliation(s)
- Kamila Bedelbaeva
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
| | - Benjamin Cameron
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
| | - John Latella
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
| | - Azamat Aslanukov
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
| | | | | | - Ellen Heber-Katz
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
- The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
4
|
Lee YL, Mathur J, Walter C, Zmuda H, Pathak A. Matrix obstructions cause multiscale disruption in collective epithelial migration by suppressing leader cell function. Mol Biol Cell 2023; 34:ar94. [PMID: 37379202 PMCID: PMC10398892 DOI: 10.1091/mbc.e22-06-0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/06/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
During disease and development, physical changes in extracellular matrix cause jamming, unjamming, and scattering in epithelial migration. However, whether disruptions in matrix topology alter collective cell migration speed and cell-cell coordination remains unclear. We microfabricated substrates with stumps of defined geometry, density, and orientation, which create obstructions for migrating epithelial cells. Here, we show that cells lose their speed and directionality when moving through densely spaced obstructions. Although leader cells are stiffer than follower cells on flat substrates, dense obstructions cause overall cell softening. Through a lattice-based model, we identify cellular protrusions, cell-cell adhesions, and leader-follower communication as key mechanisms for obstruction-sensitive collective cell migration. Our modeling predictions and experimental validations show that cells' obstruction sensitivity requires an optimal balance of cell-cell adhesions and protrusions. Both MDCK (more cohesive) and α-catenin-depleted MCF10A cells were less obstruction sensitive than wild-type MCF10A cells. Together, microscale softening, mesoscale disorder, and macroscale multicellular communication enable epithelial cell populations to sense topological obstructions encountered in challenging environments. Thus, obstruction-sensitivity could define "mechanotype" of cells that collectively migrate yet maintain intercellular communication.
Collapse
Affiliation(s)
- Ye Lim Lee
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Christopher Walter
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Hannah Zmuda
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
5
|
Walter C, Mathur J, Pathak A. Reciprocal intra- and extra-cellular polarity enables deep mechanosensing through layered matrices. Cell Rep 2023; 42:112362. [PMID: 37027304 PMCID: PMC11246724 DOI: 10.1016/j.celrep.2023.112362] [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: 09/14/2022] [Revised: 02/11/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
Adherent cells migrate on layered tissue interfaces to drive morphogenesis, wound healing, and tumor invasion. Although stiffer surfaces are known to enhance cell migration, it remains unclear whether cells sense basal stiff environments buried under softer, fibrous matrix. Using layered collagen-polyacrylamide gel systems, we unveil a migration phenotype driven by cell-matrix polarity. Here, cancer (but not normal) cells with stiff base matrix generate stable protrusions, faster migration, and greater collagen deformation because of "depth mechanosensing" through the top collagen layer. Cancer cell protrusions with front-rear polarity produce polarized collagen stiffening and deformations. Disruption of either extracellular or intracellular polarity via collagen crosslinking, laser ablation, or Arp2/3 inhibition independently abrogates depth-mechanosensitive migration of cancer cells. Our experimental findings, validated by lattice-based energy minimization modeling, present a cell migration mechanism whereby polarized cellular protrusions and contractility are reciprocated by mechanical extracellular polarity, culminating in a cell-type-dependent ability to mechanosense through matrix layers.
Collapse
Affiliation(s)
- Christopher Walter
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Amit Pathak
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
6
|
Arifianto MR, Meizikri R, Haq IBI, Susilo RI, Wahyuhadi J, Hermanto Y, Faried A. Emerging hallmark of gliomas microenvironment in evading immunity: a basic concept. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2023. [DOI: 10.1186/s41983-023-00635-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Abstract
Abstract
Background
Over the last decade, since clinical trials examining targeted therapeutics for gliomas have failed to demonstrate a meaningful increase in survival, the emphasis has recently been switched toward innovative techniques for modulating the immune response against tumors and their microenvironments (TME). Cancerous cells have eleven hallmarks which make it distinct from normal ones, among which is immune evasion. Immune evasion in glioblastoma helps it evade various treatment modalities.
Summary
Glioblastoma’s TME is composed of various array of cellular actors, ranging from peripherally derived immune cells to a variety of organ-resident specialized cell types. For example, the blood–brain barrier (BBB) serves as a selective barrier between the systemic circulation and the brain, which effectively separates it from other tissues. It is capable of blocking around 98% of molecules that transport different medications to the target tumor.
Objectives
The purpose of this paper is to offer a concise overview of fundamental immunology and how ‘clever’ gliomas avoid the immune system despite the discovery of immunotherapy for glioma.
Conclusions
Herein, we highlight the complex interplay of the tumor, the TME, and the nearby normal structures makes it difficult to grasp how to approach the tumor itself. Numerous researchers have found that the brain TME is a critical regulator of glioma growth and treatment efficacy.
Collapse
|
7
|
Krull CM, Li H, Pathak A. Nuclear export inhibition jumbles epithelial-mesenchymal states and gives rise to migratory disorder in healthy epithelia. eLife 2023; 12:e81048. [PMID: 36805020 PMCID: PMC9943065 DOI: 10.7554/elife.81048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 01/31/2023] [Indexed: 02/22/2023] Open
Abstract
Dynamic nucleocytoplasmic transport of E-M factors regulates cellular E-M states; yet, it remains unknown how simultaneously trapping these factors affects epithelia at the macroscale. To explore this question, we performed nuclear export inhibition (NEI) via leptomycin B and Selinexor treatment, which biases nuclear localization of CRM1-associated E-M factors. We examined changes in collective cellular phenotypes across a range of substrate stiffnesses. Following NEI, soft substrates elevate collective migration of MCF10A cells for up to 24 hr, while stiffer substrates reduce migration at all time points. Our results suggest that NEI disrupts migration through competition between intercellular adhesions and mechanoactivation, generally causing loss of cell-cell coordination. Specifically, across substrate stiffnesses, NEI fosters an atypical E-M state wherein MCF10A cells become both more epithelial and more mesenchymal. We observe that NEI fosters a range of these concurrent phenotypes, from more epithelial shYAP MCF10A cells to more mesenchymal MDCK II cells. α-Catenin emerges as a potential link between E-M states, where it maintains normal levels of intercellular adhesion and transmits mechanoactive characteristics to collective behavior. Ultimately, to accommodate the concurrent states observed here, we propose an expanded E-M model, which may help further understand fundamental biological phenomena and inform pathological treatments.
Collapse
Affiliation(s)
- Carly M Krull
- Department of Biomedical Engineering, Washington University in St. LouisSt LouisUnited States
| | - Haiyi Li
- Department of Computer Science and Engineering, Washington University in St. LouisSt LouisUnited States
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. LouisSt LouisUnited States
- Department of Mechanical Engineering and Materials Science, Washington University in St. LouisSt LouisUnited States
| |
Collapse
|
8
|
Daulagala AC, Kourtidis A. ECM Substrates Impact RNAi Localization at Adherens Junctions of Colon Epithelial Cells. Cells 2022; 11:3740. [PMID: 36497003 PMCID: PMC9737857 DOI: 10.3390/cells11233740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
The extracellular matrix (ECM) plays crucial roles in tissue homeostasis. Abnormalities in ECM composition are associated with pathological conditions, such as fibrosis and cancer. These ECM alterations are sensed by the epithelium and can influence its behavior through crosstalk with other mechanosensitive complexes, including the adherens junctions (AJs). We have previously shown that the AJs, through their component PLEKHA7, recruit the RNAi machinery to regulate miRNA levels and function. We have particularly shown that the junctional localization of RNAi components is critical for their function. Here, we investigated whether different ECM substrates can influence the junctional localization of RNAi complexes. To do this, we plated colon epithelial Caco2 cells on four key ECM substrates found in the colon under normal or pathogenic conditions, namely laminin, fibronectin, collagen I, and collagen IV, and we examined the subcellular distribution of PLEKHA7, and of the key RNAi components AGO2 and DROSHA. Fibronectin and collagen I negatively impacted the junctional localization of PLEKHA7, AGO2, and DROSHA when compared to laminin. Furthermore, fibronectin, collagen I, and collagen IV disrupted interactions of AGO2 and DROSHA with their essential partners GW182 and DGCR8, respectively, both at AJs and throughout the cell. Combinations of all substrates with fibronectin also negatively impacted junctional localization of PLEKHA7 and AGO2. Additionally, collagen I triggered accumulation of DROSHA at tri-cellular junctions, while both collagen I and collagen IV resulted in DROSHA accumulation at basal areas of cell-cell contact. Altogether, fibronectin and collagens I and IV, which are elevated in the stroma of fibrotic and cancerous tissues, altered localization patterns and disrupted complex formation of PLEKHA7 and RNAi components. Combined with our prior studies showing that apical junctional localization of the PLEKHA7-RNAi complex is critical for regulating tumor-suppressing miRNAs, this work points to a yet unstudied mechanism that could contribute to epithelial cell transformation.
Collapse
Affiliation(s)
| | - Antonis Kourtidis
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
9
|
Jurj A, Ionescu C, Berindan-Neagoe I, Braicu C. The extracellular matrix alteration, implication in modulation of drug resistance mechanism: friends or foes? J Exp Clin Cancer Res 2022; 41:276. [PMID: 36114508 PMCID: PMC9479349 DOI: 10.1186/s13046-022-02484-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/01/2022] [Indexed: 11/10/2022] Open
Abstract
The extracellular matrix (ECM) is an important component of the tumor microenvironment (TME), having several important roles related to the hallmarks of cancer. In cancer, multiple components of the ECM have been shown to be altered. Although most of these alterations are represented by the increased or decreased quantity of the ECM components, changes regarding the functional alteration of a particular ECM component or of the ECM as a whole have been described. These alterations can be induced by the cancer cells directly or by the TME cells, with cancer-associated fibroblasts being of particular interest in this regard. Because the ECM has this wide array of functions in the tumor, preclinical and clinical studies have assessed the possibility of targeting the ECM, with some of them showing encouraging results. In the present review, we will highlight the most relevant ECM components presenting a comprehensive description of their physical, cellular and molecular properties which can alter the therapy response of the tumor cells. Lastly, some evidences regarding important biological processes were discussed, offering a more detailed understanding of how to modulate altered signalling pathways and to counteract drug resistance mechanisms in tumor cells.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania
| | - Calin Ionescu
- 7Th Surgical Department, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012, Cluj-Napoca, Romania
- Surgical Department, Municipal Hospital, 400139, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
- Research Center for Oncopathology and Translational Medicine (CCOMT), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, 540139, Targu Mures, Romania.
| |
Collapse
|
10
|
Flick Jaecker F, Almeida JA, Krull CM, Pathak A. Nucleoli in epithelial cell collectives respond to tumorigenic, spatial, and mechanical cues. Mol Biol Cell 2022; 33:br19. [PMID: 35830599 PMCID: PMC9582805 DOI: 10.1091/mbc.e22-02-0070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Cancer cells are known to have larger nucleoli, consistent with their higher transcriptional and translational demands. Meanwhile, on stiff extracellular matrix, normal epithelial cells can exhibit genomic and proteomic mechanoactivation toward tumorigenic transformations, such as epithelial-mesenchymal transition and enhanced migration. However, while nucleolar bodies regulate the protein synthesis required for mechanosensation, it remains unknown whether mechanical and spatial extracellular cues can in turn alter nucleoli. Here, we culture mammary epithelial cell sheets on matrices of varying stiffness and show that cancer cells have more nucleoli, with nucleoli occupying larger areas compared with normal cells. By contrast, within normal epithelial sheets, stiffer matrices and leader positioning of cells induce larger nucleolar areas and more nucleolar bodies over time. The observed leader-follower nucleolar differences stem from distinct rates of cell cycle progression. In the nucleoplasm, leader cells on stiffer matrices exhibit higher heterochromatin marker expression and DNA compaction around nucleolar bodies. Overall, our findings advance the emerging framework of cellular mechanobiology in which mechanical cues from the extracellular matrix transmit into the nucleoplasm to alter nucleolar composition, potentially resulting in mechanosensitive ribosomal biogenesis. Ultimately, this proposed mechanosensitivity of nucleoli and associated protein synthesis could have wide implications in disease, development, and regeneration.
Collapse
Affiliation(s)
| | - José A Almeida
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Carly M Krull
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Amit Pathak
- Department of Mechanical Engineering & Materials Science and.,Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| |
Collapse
|
11
|
Tian H, Shi H, Yu J, Ge S, Ruan J. Biophysics Role and Biomimetic Culture Systems of ECM Stiffness in Cancer EMT. GLOBAL CHALLENGES (HOBOKEN, NJ) 2022; 6:2100094. [PMID: 35712024 PMCID: PMC9189138 DOI: 10.1002/gch2.202100094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/14/2022] [Indexed: 06/15/2023]
Abstract
Oncological diseases have become the second leading cause of death from noncommunicable diseases worldwide and a major threat to human health. With the continuous progress in cancer research, the mechanical cues from the tumor microenvironment environment (TME) have been found to play an irreplaceable role in the progression of many cancers. As the main extracellular mechanical signal carrier, extracellular matrix (ECM) stiffness may influence cancer progression through biomechanical transduction to modify downstream gene expression, promote epithelial-mesenchymal transition (EMT), and regulate the stemness of cancer cells. EMT is an important mechanism that induces cancer cell metastasis and is closely influenced by ECM stiffness, either independently or in conjunction with other molecules. In this review, the unique role of ECM stiffness in EMT in different kinds of cancers is first summarized. By continually examining the significance of ECM stiffness in cancer progression, a biomimetic culture system based on 3D manufacturing and novel material technologies is developed to mimic ECM stiffness. The authors then look back on the novel development of the ECM stiffness biomimetic culture systems and finally provide new insights into ECM stiffness in cancer progression which can broaden the fields' horizons with a view toward developing new cancer diagnosis methods and therapies.
Collapse
Affiliation(s)
- Hao Tian
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| | - Hanhan Shi
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| | - Jie Yu
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| | - Shengfang Ge
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| | - Jing Ruan
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| |
Collapse
|
12
|
Speer JE, Barcellona MN, Lu MY, Zha Z, Jing L, Gupta MC, Buchowski JM, Kelly MP, Setton LA. Development of a library of laminin-mimetic peptide hydrogels for control of nucleus pulposus cell behaviors. J Tissue Eng 2021; 12:20417314211021220. [PMID: 34188794 PMCID: PMC8211742 DOI: 10.1177/20417314211021220] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022] Open
Abstract
The nucleus pulposus (NP) of the intervertebral disc plays a critical role in
distributing mechanical loads to the axial skeleton. Alterations in NP cells and,
consequently, NP matrix are some of the earliest changes in the development of disc
degeneration. Previous studies demonstrated a role for laminin-presenting biomaterials in
promoting a healthy phenotype for human NP cells from degenerated tissue. Here we
investigate the use of laminin-mimetic peptides presented individually or in combination
on a poly(ethylene) glycol hydrogel as a platform to modulate the behaviors of
degenerative human NP cells. Data confirm that NP cells attach to select laminin-mimetic
peptides that results in cell signaling downstream of integrin and syndecan binding.
Furthermore, the peptide-functionalized hydrogels demonstrate an ability to promote cell
behaviors that mimic that of full-length laminins. These results identify a set of
peptides that can be used to regulate NP cell behaviors toward a regenerative engineering
strategy.
Collapse
Affiliation(s)
- Julie E Speer
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Marcos N Barcellona
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael Y Lu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Zizhen Zha
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Liufang Jing
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Munish C Gupta
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacob M Buchowski
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael P Kelly
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori A Setton
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
13
|
Liu Z, Sun D, Zhu Q, Liu X. The screening of immune-related biomarkers for prognosis of lung adenocarcinoma. Bioengineered 2021; 12:1273-1285. [PMID: 33870858 PMCID: PMC8806236 DOI: 10.1080/21655979.2021.1911211] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lung adenocarcinoma (LUAD) accounts for a frequently seen non-small cell lung cancer (NSCLC) histological subtype, and it is associated with dismal prognostic outcome. However, the benefits of traditional treatment are still limited, and the efficacies of immunotherapy are quite different. Therefore, it is of great significance to identify novel immune-related therapeutic targets in lung adenocarcinoma. In this study, we identified a set of immune-related biomarkers for prognosis of lung adenocarcinoma, which could provide new ideas for immunotherapy of lung adenocarcinoma. Datasets related to LUAD were filtered from the GEO database. The appropriate packages were used to identify differentially expressed genes (DEGs) and to carry out enrichment analysis, followed by the construction of prognostic biomarkers. The Kaplan-Meier (K-M) curves were plotted to analyze patient survival based on hub genes. Associations between the expression of selected biomarkers and six types of tumor-infiltrating immune cells were evaluated based on the online tool TIMER. After analyzing five GEO datasets(GSE32867, GSE46539, GSE63459, GSE75037 and GSE116959), we discovered altogether 67 DEGs, among which, 15 showed up-regulation while 52 showed down-regulation. Enrichments of integrated DEGs were identified in the ontology categories. CAV1, CFD, FMO2 and CLEC3B were eventually selected as independent prognostic biomarkers, they were correlated with clinical outcomes of LUAD patients. Moreover, a positive correlation was observed between biomarker expression and all different types of immune infiltration, and the expression level of the four biomarkers was all positively related to macrophage.
Collapse
Affiliation(s)
- Zhonghui Liu
- Department of Geriatrics, Peking University First Hospital, Beijing, China
| | - Dan Sun
- Department of Geriatrics, Peking University First Hospital, Beijing, China
| | - Qing Zhu
- Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xinmin Liu
- Department of Geriatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
14
|
Patil VA, Masters KS. Engineered Collagen Matrices. Bioengineering (Basel) 2020; 7:E163. [PMID: 33339157 PMCID: PMC7765577 DOI: 10.3390/bioengineering7040163] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 01/10/2023] Open
Abstract
Collagen is the most abundant protein in mammals, accounting for approximately one-third of the total protein in the human body. Thus, it is a logical choice for the creation of biomimetic environments, and there is a long history of using collagen matrices for various tissue engineering applications. However, from a biomaterial perspective, the use of collagen-only scaffolds is associated with many challenges. Namely, the mechanical properties of collagen matrices can be difficult to tune across a wide range of values, and collagen itself is not highly amenable to direct chemical modification without affecting its architecture or bioactivity. Thus, many approaches have been pursued to design scaffold environments that display critical features of collagen but enable improved tunability of physical and biological characteristics. This paper provides a brief overview of approaches that have been employed to create such engineered collagen matrices. Specifically, these approaches include blending of collagen with other natural or synthetic polymers, chemical modifications of denatured collagen, de novo creation of collagen-mimetic chains, and reductionist methods to incorporate collagen moieties into other materials. These advancements in the creation of tunable, engineered collagen matrices will continue to enable the interrogation of novel and increasingly complex biological questions.
Collapse
Affiliation(s)
| | - Kristyn S. Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA;
| |
Collapse
|
15
|
Abstract
Lung cancer is the world's most common malignancies and ranks first among all cancer-related deaths. Lung adenocarcinoma (LUAD) is the most frequent histological type in lung cancer. Its pathogenesis has not yet been fully elucidated, so it is of great significance to explore related genes for elucidating the molecular mechanism involved in occurrence and development of LUAD.To explore the crucial genes associated with LUAD development and progression, microarray datasets GSE7670, GSE10072, and GSE31547 were acquired from the Gene Expression Omnibus (GEO) database. R language Limma package was adopted to screen the differentially expressed genes (DEGs). The clusterProfiler package was used for enrichment analysis and annotation of the Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) pathways for DEGs. The Search Tool for the Retrieval of Interacting Genes database (STRING) was used to construct the protein interaction network for DEGs, while Cytoscape was adopted to visualize it. The functional module was screened with Cytoscape's MCODE (The Molecular Complex Detection) plugin. The crucial genes associated with LUAD were identified by cytoHubba plugin. Kaplan-Meier plotter online tool was used to perform survival analysis of the hub gene.Three hundred twenty-one DEGs in total were screened, of which 105 were upregulated and 216 were downregulated. It was found that some GO terms and pathways (e.g., collagen trimer, extracellular structure organization, heparin binding, complement and coagulation cascades, malaria, protein digestion and absorption, and PPAR signaling pathway) were considerably enriched in DEGs. UBE2C, TOP2A, RRM2, CDC20, CCNB2, KIAA0101, BUB1B, TPX2, PRC1, and CDK1 were identified as crucial genes. Survival analysis showed that the overexpression of UBE2C, TOP2A, RRM2, CDC20, CCNB2, KIAA0101, BUB1B, TPX2, and PRC1 significantly reduced the overall survival of LUAD patients. One of the crucial genes: UBE2C was validated by immunohistochemistry to be upregulated in LUAD tissues.This study screened out potential biomarkers of LUAD, providing a theoretical basis for elucidating the pathogenesis and evaluating the prognosis of LUAD.
Collapse
|
16
|
Henke E, Nandigama R, Ergün S. Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy. Front Mol Biosci 2020; 6:160. [PMID: 32118030 PMCID: PMC7025524 DOI: 10.3389/fmolb.2019.00160] [Citation(s) in RCA: 660] [Impact Index Per Article: 132.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Solid tumors are complex organ-like structures that consist not only of tumor cells but also of vasculature, extracellular matrix (ECM), stromal, and immune cells. Often, this tumor microenvironment (TME) comprises the larger part of the overall tumor mass. Like the other components of the TME, the ECM in solid tumors differs significantly from that in normal organs. Intratumoral signaling, transport mechanisms, metabolisms, oxygenation, and immunogenicity are strongly affected if not controlled by the ECM. Exerting this regulatory control, the ECM does not only influence malignancy and growth of the tumor but also its response toward therapy. Understanding the particularities of the ECM in solid tumor is necessary to develop approaches to interfere with its negative effect. In this review, we will also highlight the current understanding of the physical, cellular, and molecular mechanisms by which the pathological tumor ECM affects the efficiency of radio-, chemo-, and immunotherapy. Finally, we will discuss the various strategies to target and modify the tumor ECM and how they could be utilized to improve response to therapy.
Collapse
Affiliation(s)
- Erik Henke
- Department of Medicine, Institute of Anatomy and Cell Biology, Universität Würzburg, Würzburg, Germany
| | - Rajender Nandigama
- Department of Medicine, Institute of Anatomy and Cell Biology, Universität Würzburg, Würzburg, Germany
| | - Süleyman Ergün
- Department of Medicine, Institute of Anatomy and Cell Biology, Universität Würzburg, Würzburg, Germany
| |
Collapse
|
17
|
Sarker B, Bagchi A, Walter C, Almeida J, Pathak A. Longer collagen fibers trigger multicellular streaming on soft substrates via enhanced forces and cell-cell cooperation. J Cell Sci 2019; 132:jcs226753. [PMID: 31444287 PMCID: PMC6765186 DOI: 10.1242/jcs.226753] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
Grouped cells often leave large cell colonies in the form of narrow multicellular streams. However, it remains unknown how collective cell streaming exploits specific matrix properties, like stiffness and fiber length. It is also unclear how cellular forces, cell-cell adhesion and velocities are coordinated within streams. To independently tune stiffness and collagen fiber length, we developed new hydrogels and discovered invasion-like streaming of normal epithelial cells on soft substrates coated with long collagen fibers. Here, streams arise owing to a surge in cell velocities, forces, YAP activity and expression of mesenchymal marker proteins in regions of high-stress anisotropy. Coordinated velocities and symmetric distribution of tensile and compressive stresses support persistent stream growth. Stiff matrices diminish cell-cell adhesions, disrupt front-rear velocity coordination and do not promote sustained fiber-dependent streaming. Rac inhibition reduces cell elongation and cell-cell cooperation, resulting in a complete loss of streaming in all matrix conditions. Our results reveal a stiffness-modulated effect of collagen fiber length on collective cell streaming and unveil a biophysical mechanism of streaming governed by a delicate balance of enhanced forces, monolayer cohesion and cell-cell cooperation.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Bapi Sarker
- Department of Mechanical Engineering & Materials Science, Washington University, St Louis, MO 63130, USA
| | - Amrit Bagchi
- Department of Mechanical Engineering & Materials Science, Washington University, St Louis, MO 63130, USA
| | - Christopher Walter
- Department of Biomedical Engineering, Washington University, St Louis, MO 63130, USA
| | - José Almeida
- Department of Biomedical Engineering, Washington University, St Louis, MO 63130, USA
| | - Amit Pathak
- Department of Mechanical Engineering & Materials Science, Washington University, St Louis, MO 63130, USA
- Department of Biomedical Engineering, Washington University, St Louis, MO 63130, USA
| |
Collapse
|
18
|
Perry G, Xiao W, Welsh GI, Perriman AW, Lennon R. Engineered basement membranes: from in vivo considerations to cell-based assays. Integr Biol (Camb) 2018; 10:680-695. [DOI: 10.1039/c8ib00138c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Engineered basement membranes are required to mimic in vivo properties within cell-based assays.
Collapse
Affiliation(s)
- Guillaume Perry
- Sorbonne Université, Laboratoire d’Electronique et d’Electromagnétisme
- F-75005 Paris
- France
| | - Wenjin Xiao
- School of Cellular and Molecular Medicine, University of Bristol
- BS8 1TD Bristol
- UK
| | - Gavin I. Welsh
- Bristol Renal, Bristol Medical School, University of Bristol
- BS1 3NY Bristol
- UK
| | - Adam W. Perriman
- School of Cellular and Molecular Medicine, University of Bristol
- BS8 1TD Bristol
- UK
| | - Rachel Lennon
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester
- M13 9PT Manchester
- UK
| |
Collapse
|