1
|
Srinivasan S, Ramos-Lewis W, Morais MR, Chi Q, Soh AW, Williams E, Lennon R, Sherwood DR. A collagen IV fluorophore knock-in toolkit reveals trimer diversity in C. elegans basement membranes. J Cell Biol 2025; 224:e202412118. [PMID: 40100062 PMCID: PMC11917169 DOI: 10.1083/jcb.202412118] [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: 12/14/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
The type IV collagen triple helix, composed of three ⍺-chains, is a core basement membrane (BM) component that assembles into a network within BMs. Endogenous tagging of all ⍺-chains with genetically encoded fluorophores has remained elusive, limiting our understanding of this crucial BM component. Through genome editing, we show that the C termini of the C. elegans type IV collagen ⍺-chains EMB-9 and LET-2 can be fused to a variety of fluorophores to create a strain toolkit with wild-type health. Using quantitative imaging, our results suggest a preference for LET-2-LET-2-EMB-9 trimer construction, but also tissue-specific flexibility in trimers assembled driven by differences in ⍺-chain expression levels. By tagging emb-9 and let-2 mutants that model human Gould syndrome, a complex multitissue disorder, we further discover defects in extracellular accumulation and turnover that might help explain disease pathology. Together, our findings identify a permissive tagging site in C. elegans that will allow diverse studies on type IV collagen regulation and function in animals.
Collapse
Affiliation(s)
| | | | - Mychel R.P.T. Morais
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Qiuyi Chi
- Department of Biology, Duke University, Durham, NC, USA
| | - Adam W.J. Soh
- Department of Biology, Duke University, Durham, NC, USA
| | - Emily Williams
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Rachel Lennon
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | | |
Collapse
|
2
|
Srinivasan S, Sherwood DR. The life cycle of type IV collagen. Matrix Biol 2025:S0945-053X(25)00037-X. [PMID: 40306374 DOI: 10.1016/j.matbio.2025.04.004] [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: 03/31/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Type IV collagen is a large triple helical molecule that forms a covalently cross-linked network within basement membranes (BMs). Type IV collagen networks play key roles in mechanically supporting tissues, shaping organs, filtering blood, and cell signaling. To ensure tissue health and function, all aspects of the type IV collagen life cycle must be carried out accurately. However, the large triple helical structure and complex life-cycle of type IV collagen, poses many challenges to cells and tissues. Type IV collagen predominantly forms heterotrimers and to ensure proper construction, expression of the distinct α-chains that comprise a heterotrimer needs tight regulation. The α-chains must also be accurately modified by several enzymes, some of which are specific to collagens, to build and stabilize the triple helical trimer. In addition, type IV collagen is exceptionally long (400nm) and thus the packaging and trafficking of the triple helical trimer from the ER to the Golgi must be modified to accommodate the large type IV collagen molecule. During ER-to-Golgi trafficking, as well as during secretion and transport in the extracellular space type IV collagen also associates with specific chaperone molecules that maintain the structure and solubility of collagen IV. Type IV collagen trimers are then delivered to BMs from local and distant sources where they are integrated into BMs by interactions with cell surface receptors and many diverse BM resident proteins. Within BMs type IV collagen self-associates into a network and is crosslinked by BM resident enzymes. Finally, homeostatic type IV collagen levels in BMs are maintained by poorly understood mechanisms involving proteolysis and endocytosis. Here, we provide an overview of the life cycle of collagen IV, highlighting unique mechanisms and poorly understood aspects of type IV collagen regulation.
Collapse
Affiliation(s)
- Sandhya Srinivasan
- Department of Biology, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - David R Sherwood
- Department of Biology, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA.
| |
Collapse
|
3
|
Delage S, Zadhoosh A, You W, Brown TJ, Ringuette MJ. Drosophila SPARC collagen IV chaperone-like activity essential for development is unique to the fat body. iScience 2025; 28:112111. [PMID: 40241767 PMCID: PMC12002606 DOI: 10.1016/j.isci.2025.112111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/02/2024] [Accepted: 02/21/2025] [Indexed: 04/18/2025] Open
Abstract
Drosophila fat body-derived SPARC acts as a chaperone for collagen IV (Col(IV)), enabling their diffusion and incorporation into distal tissue basement membranes (BMs). Disruption of SPARC or Col(IV) production by the fat body is lethal, despite expression by other tissues such as imaginal discs. Wing disc-derived SPARC does not associate with Col(IV) in BMs and is not essential for survival. We show that differential association of fat body- and wing disc-derived SPARC with Col(IV) is not due to differences in SPARC glycosylation nor to the absence of SPARC and Col(IV) co-expression. Further, we demonstrate that SPARC domain II/III produced by the fat body is sufficient for Col(IV) diffusion to both proximal and distal BMs, and rescues lethality associated with loss of SPARC. However, SPARC domain II/III does not diffuse beyond the hemolymph. Thus, the essential Col(IV) chaperone-like activity specific to fat body-derived SPARC is not required beyond the hemolymph.
Collapse
Affiliation(s)
- Samuel Delage
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Arya Zadhoosh
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - William You
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Theodore Joseph Brown
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON M5G 1E2, Canada
- Lunenfeld-Tanenbaum, Research Institute at Sinai Health Systems, Toronto, ON M5G 1X5, Canada
| | | |
Collapse
|
4
|
Brigant B, Metzinger-Le Meuth V, Boyartchuk V, Ouled-Haddou H, Guerrera IC, Rochette J, Metzinger L. A proteomic study of the downregulation of TRIM37 on chondrocytes: Implications for the MULIBREY syndrome. Bone 2024; 187:117205. [PMID: 39019132 DOI: 10.1016/j.bone.2024.117205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
MULIBREY nanism which results from autosomal recessive mutations in TRIM37 impacts skeletal development, leading to growth delay with complications in multiple organs. In this study, we employed a combined proteomics and qPCR screening approach to investigate the molecular alterations in the CHON-002 cell line by comparing CHON-002 wild-type (WT) cells to CHON-002 TRIM37 knockdown (KD) cells. Our proteomic analysis demonstrated that TRIM37 depletion predominantly affects the expression of extracellular matrix proteins (ECM). Specifically, nanoLC-MS/MS experiments revealed an upregulation of SPARC, and collagen products (COL1A1, COL3A1, COL5A1) in response to TRIM37 KD. Concurrently, large-scale qPCR assays targeting osteogenesis-related genes corroborated these dysregulations of SPARC at the mRNA level. Gene ontology enrichment analysis highlighted the involvement of dysregulated proteins in ECM organization and TGF-β signaling pathways, indicating a role for TRIM37 in maintaining ECM integrity and regulating chondrocyte proliferation. These findings suggest that TRIM37 deficiency in chondrocytes change ECM protein composition and could impairs long bone growth, contributing to the pathophysiology of MULIBREY nanism.
Collapse
Affiliation(s)
- Benjamin Brigant
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, 80000 Amiens, France; Centre of Molecular Inflammation Research (CEMIR), Department of Clinical Research and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences (MH), Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Valérie Metzinger-Le Meuth
- INSERM UMRS 1148, Laboratory for Vascular Translational Science (LVTS), UFR SMBH, University of Sorbonne Paris Nord, 93000 Bobigny, France
| | - Victor Boyartchuk
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical Research and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences (MH), Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Surgery Clinic, St. Olav's Hospital HF, Trondheim, Norway; Centre for Integrative Genetics, Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Hakim Ouled-Haddou
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, 80000 Amiens, France
| | - Ida Chiara Guerrera
- Proteomics Platform Necker, Université Paris Cité-Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, 75015, Paris, France
| | - Jacques Rochette
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, 80000 Amiens, France
| | - Laurent Metzinger
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, 80000 Amiens, France.
| |
Collapse
|
5
|
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
|
6
|
Jones RA, Trejo B, Sil P, Little KA, Pasolli HA, Joyce B, Posfai E, Devenport D. An mTurq2-Col4a1 mouse model allows for live visualization of mammalian basement membrane development. J Cell Biol 2024; 223:e202309074. [PMID: 38051393 PMCID: PMC10697824 DOI: 10.1083/jcb.202309074] [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: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
Basement membranes (BMs) are specialized sheets of extracellular matrix that underlie epithelial and endothelial tissues. BMs regulate the traffic of cells and molecules between compartments, and participate in signaling, cell migration, and organogenesis. The dynamics of mammalian BMs, however, are poorly understood, largely due to a lack of models in which core BM components are endogenously labeled. Here, we describe the mTurquoise2-Col4a1 mouse in which we fluorescently tag collagen IV, the main component of BMs. Using an innovative planar-sagittal live imaging technique to visualize the BM of developing skin, we directly observe BM deformation during hair follicle budding and basal progenitor cell divisions. The BM's inherent pliability enables dividing cells to remain attached to and deform the BM, rather than lose adhesion as generally thought. Using FRAP, we show BM collagen IV is extremely stable, even during periods of rapid epidermal growth. These findings demonstrate the utility of the mTurq2-Col4a1 mouse to shed new light on mammalian BM developmental dynamics.
Collapse
Affiliation(s)
- Rebecca A. Jones
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Brandon Trejo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Parijat Sil
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - H. Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - Bradley Joyce
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
7
|
Jones RA, Trejo B, Sil P, Little KA, Pasolli HA, Joyce B, Posfai E, Devenport D. A Window into Mammalian Basement Membrane Development: Insights from the mTurq2-Col4a1 Mouse Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559396. [PMID: 37808687 PMCID: PMC10557719 DOI: 10.1101/2023.09.27.559396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Basement membranes (BMs) are specialized sheets of extracellular matrix that underlie epithelial and endothelial tissues. BMs regulate traffic of cells and molecules between compartments, and participate in signaling, cell migration and organogenesis. The dynamics of mammalian BMs, however, are poorly understood, largely due to a lack of models in which core BM components are endogenously labelled. Here, we describe the mTurquoise2-Col4a1 mouse, in which we fluorescently tag collagen IV, the main component of BMs. Using an innovative Planar-Sagittal live imaging technique to visualize the BM of developing skin, we directly observe BM deformation during hair follicle budding and basal progenitor cell divisions. The BM's inherent pliability enables dividing cells to remain attached to and deform the BM, rather than lose adhesion as generally thought. Using FRAP, we show BM collagen IV is extremely stable, even during periods of rapid epidermal growth. These findings demonstrate the utility of the mTurq2-Col4a1 mouse to shed new light on mammalian BM developmental dynamics.
Collapse
Affiliation(s)
- Rebecca A Jones
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Brandon Trejo
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Parijat Sil
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Katherine A Little
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - H Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, 1230 York Ave., New York, NY 10065
| | - Bradley Joyce
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
8
|
Bian C, Zheng Z, Su J, Chang S, Yu H, Bao J, Xin Y, Jiang X. Copper homeostasis and cuproptosis in tumor pathogenesis and therapeutic strategies. Front Pharmacol 2023; 14:1271613. [PMID: 37767404 PMCID: PMC10520736 DOI: 10.3389/fphar.2023.1271613] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Copper is an indispensable micronutrient for the development and replication of all eukaryotes, and its redox properties are both harmful and beneficial to cells. An imbalance in copper homeostasis is thought to be involved in carcinogenesis. Importantly, cancer cell proliferation, angiogenesis, and metastasis cannot be separated from the effects of copper. Cuproposis is a copper-dependent form of cell death that differs from other existing modalities of regulatory cell death. The role of cuproptosis in the pathogenesis of the nervous and cardiovascular systems has been widely studied; however, its impact on malignant tumors is yet to be fully understood from a clinical perspective. Exploring signaling pathways related to cuproptosis will undoubtedly provide a new perspective for the development of anti-tumor drugs in the future. Here, we systematically review the systemic and cellular metabolic processes of copper and the regulatory mechanisms of cuproptosis in cancer. In addition, we discuss the possibility of targeting copper ion drugs to prolong the survival of cancer patients, with an emphasis on the most representative copper ionophores and chelators. We suggest that attention should be paid to the potential value of copper in the treatment of specific cancers.
Collapse
Affiliation(s)
- Chenbin Bian
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Zhuangzhuang Zheng
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Jing Su
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Sitong Chang
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Huiyuan Yu
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Jindian Bao
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| |
Collapse
|
9
|
Zhou C, Yang J, Liu T, Jia R, Yang L, Sun P, Zhao W. Copper metabolism and hepatocellular carcinoma: current insights. Front Oncol 2023; 13:1186659. [PMID: 37476384 PMCID: PMC10355993 DOI: 10.3389/fonc.2023.1186659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023] Open
Abstract
Copper is an essential trace element that acts as a cofactor in various enzyme active sites in the human body. It participates in numerous life activities, including lipid metabolism, energy metabolism, and neurotransmitter synthesis. The proposal of "Cuproptosis" has made copper metabolism-related pathways a research hotspot in the field of tumor therapy, which has attracted great attention. This review discusses the biological processes of copper uptake, transport, and storage in human cells. It highlights the mechanisms by which copper metabolism affects hepatocellular carcinogenesis and metastasis, including autophagy, apoptosis, vascular invasion, cuproptosis, and ferroptosis. Additionally, it summarizes the current clinical applications of copper metabolism-related drugs in antitumor therapy.
Collapse
Affiliation(s)
- Cheng Zhou
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinqiu Yang
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tong Liu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ran Jia
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lin Yang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital Affiliated to Shaanxi University of Chinese Medicine, Xianyang, China
| | - Pengfei Sun
- Department of Orthopaedics, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wenxia Zhao
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
10
|
Wang M, Ji C, Zhang Y, Zhang Z, Zhang Y, Guo H, Qiao N, Zhou X, Cao X, Ye Z, Yu Y, Melnikov V, Gong W, He M, Zhang Z, Zhao Y, Wang X, Wei G, Ye Z. Global changes in chromatin accessibility and transcription in growth hormone-secreting pituitary adenoma. Endocrine 2022; 78:329-342. [PMID: 35947334 PMCID: PMC9584994 DOI: 10.1007/s12020-022-03155-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Growth hormone-secreting pituitary adenoma (GHPA) is an insidious disease with persistent hypersecretion of growth hormone and insulin-like growth factor 1, causing increased morbidity and mortality. Previous studies have investigated the transcription of GHPA. However, the gene regulatory landscape has not been fully characterized. The objective of our study was to unravel the changes in chromatin accessibility and transcription in GHPA. METHODS Six patients diagnosed with GHPA in the Department of Neurosurgery at Huashan Hospital were enrolled in our study. Primary pituitary adenoma tissues and adjacent normal pituitary specimens with no morphologic abnormalities from these six patients were obtained at surgery. RNA sequencing (RNA-seq) and assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) were applied to investigate the underlying relationship between gene expression and chromatin accessibility changes in GHPA. RESULTS Totally, 1528 differential expression genes (DEGs) were identified by transcriptomics analyses, including 725 up-regulated and 803 down-regulated. Further, we obtained 64 significantly DEGs including 10 DEGs were elevated and 54 DEGs were negligibly expressed in tumors tissues. The up-regulated DEGs were mainly involved in terms related to synapse formation, nervous system development and secretory pathway. In parallel, 3916 increased and 2895 decreased chromatin-accessible regions were mapped by ATAC-seq. Additionally, the chromatin accessible changes were frequently located adjacent to transcription factor CTCF and Rfx2 binding site. CONCLUSIONS Our results are the first to demonstrate the landscape of chromatin accessibility in GHPA, which may contribute to illustrate the underlying transcriptional regulation mechanism of this disease.
Collapse
Affiliation(s)
- Meng Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenxing Ji
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yichao Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Zhiqiang Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huiping Guo
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Nidan Qiao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Xiang Zhou
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Xiaoyun Cao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Zhen Ye
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yifei Yu
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Vladimir Melnikov
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Gong
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min He
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhaoyun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Sate Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Neurosurgical Institute of Fudan University, Fudan University, Shanghai, China
| | - Xuelong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Gang Wei
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Zhao Ye
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| |
Collapse
|
11
|
Memo1 binds reduced copper ions, interacts with copper chaperone Atox1, and protects against copper-mediated redox activity in vitro. Proc Natl Acad Sci U S A 2022; 119:e2206905119. [PMID: 36067318 PMCID: PMC9477392 DOI: 10.1073/pnas.2206905119] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Since many proteins depend on copper ions for functionality, it is not surprising that cancer cells have a high demand for copper. Still, free copper ions are toxic as they can potentially catalyze the formation of harmful reactive oxygen species (ROS) upon coupling redox cycling between Cu(I) and Cu(II) with reduction of O2. Here, we investigated copper binding to Memo1, an oncogenic protein linked to cancer. We demonstrate that Memo1 coordinates reduced but not oxidized copper ions, thereby preventing the copper ions from acting as redox catalysts for ROS generation. As Memo1 is a putative target for the treatment of cancer, it is of importance to identify its binding partners (e.g., metal ions) and the functional consequences of such interactions. The protein mediator of ERBB2-driven cell motility 1 (Memo1) is connected to many signaling pathways that play key roles in cancer. Memo1 was recently postulated to bind copper (Cu) ions and thereby promote the generation of reactive oxygen species (ROS) in cancer cells. Since the concentration of Cu as well as ROS are increased in cancer cells, both can be toxic if not well regulated. Here, we investigated the Cu-binding capacity of Memo1 using an array of biophysical methods at reducing as well as oxidizing conditions in vitro. We find that Memo1 coordinates two reduced Cu (Cu(I)) ions per protein, and, by doing so, the metal ions are shielded from ROS generation. In support of biological relevance, we show that the cytoplasmic Cu chaperone Atox1, which delivers Cu(I) in the secretory pathway, can interact with and exchange Cu(I) with Memo1 in vitro and that the two proteins exhibit spatial proximity in breast cancer cells. Thus, Memo1 appears to act as a Cu(I) chelator (perhaps shuttling the metal ion to Atox1 and the secretory path) that protects cells from Cu-mediated toxicity, such as uncontrolled formation of ROS. This Memo1 functionality may be a safety mechanism to cope with the increased demand of Cu ions in cancer cells.
Collapse
|
12
|
Bordini M, Soglia F, Davoli R, Zappaterra M, Petracci M, Meluzzi A. Molecular Pathways and Key Genes Associated With Breast Width and Protein Content in White Striping and Wooden Breast Chicken Pectoral Muscle. Front Physiol 2022; 13:936768. [PMID: 35874513 PMCID: PMC9304951 DOI: 10.3389/fphys.2022.936768] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/17/2022] [Indexed: 01/10/2023] Open
Abstract
Growth-related abnormalities affecting modern chickens, known as White Striping (WS) and Wooden Breast (WB), have been deeply investigated in the last decade. Nevertheless, their precise etiology remains unclear. The present study aimed at providing new insights into the molecular mechanisms involved in their onset by identifying clusters of co-expressed genes (i.e., modules) and key loci associated with phenotypes highly related to the occurrence of these muscular disorders. The data obtained by a Weighted Gene Co-expression Network Analysis (WGCNA) were investigated to identify hub genes associated with the parameters breast width (W) and total crude protein content (PC) of Pectoralis major muscles (PM) previously harvested from 12 fast-growing broilers (6 normal vs. 6 affected by WS/WB). W and PC can be considered markers of the high breast yield of modern broilers and the impaired composition of abnormal fillets, respectively. Among the identified modules, the turquoise (r = -0.90, p < 0.0001) and yellow2 (r = 0.91, p < 0.0001) were those most significantly related to PC and W, and therefore respectively named “protein content” and “width” modules. Functional analysis of the width module evidenced genes involved in the ubiquitin-mediated proteolysis and inflammatory response. GTPase activator activity, PI3K-Akt signaling pathway, collagen catabolic process, and blood vessel development have been detected among the most significant functional categories of the protein content module. The most interconnected hub genes detected for the width module encode for proteins implicated in the adaptive responses to oxidative stress (i.e., THRAP3 and PRPF40A), and a member of the inhibitor of apoptosis family (i.e., BIRC2) involved in contrasting apoptotic events related to the endoplasmic reticulum (ER)-stress. The protein content module showed hub genes coding for different types of collagens (such as COL6A3 and COL5A2), along with MMP2 and SPARC, which are implicated in Collagen type IV catabolism and biosynthesis. Taken together, the present findings suggested that an ER stress condition may underly the inflammatory responses and apoptotic events taking place within affected PM muscles. Moreover, these results support the hypothesis of a role of the Collagen type IV in the cascade of events leading to the occurrence of WS/WB and identify novel actors probably involved in their onset.
Collapse
Affiliation(s)
- Martina Bordini
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum—University of Bologna, Bologna, Italy
| | - Francesca Soglia
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum—University of Bologna, Cesena, Italy
| | - Roberta Davoli
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum—University of Bologna, Bologna, Italy
| | - Martina Zappaterra
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum—University of Bologna, Bologna, Italy
- *Correspondence: Martina Zappaterra,
| | - Massimiliano Petracci
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum—University of Bologna, Cesena, Italy
| | - Adele Meluzzi
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum—University of Bologna, Bologna, Italy
| |
Collapse
|
13
|
Cote LE, Feldman JL. Won't You be My Neighbor: How Epithelial Cells Connect Together to Build Global Tissue Polarity. Front Cell Dev Biol 2022; 10:887107. [PMID: 35800889 PMCID: PMC9253303 DOI: 10.3389/fcell.2022.887107] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial tissues form continuous barriers to protect against external environments. Within these tissues, epithelial cells build environment-facing apical membranes, junction complexes that anchor neighbors together, and basolateral surfaces that face other cells. Critically, to form a continuous apical barrier, neighboring epithelial cells must align their apico-basolateral axes to create global polarity along the entire tissue. Here, we will review mechanisms of global tissue-level polarity establishment, with a focus on how neighboring epithelial cells of different origins align their apical surfaces. Epithelial cells with different developmental origins and/or that polarize at different times and places must align their respective apico-basolateral axes. Connecting different epithelial tissues into continuous sheets or tubes, termed epithelial fusion, has been most extensively studied in cases where neighboring cells initially dock at an apical-to-apical interface. However, epithelial cells can also meet basal-to-basal, posing several challenges for apical continuity. Pre-existing basement membrane between the tissues must be remodeled and/or removed, the cells involved in docking are specialized, and new cell-cell adhesions are formed. Each of these challenges can involve changes to apico-basolateral polarity of epithelial cells. This minireview highlights several in vivo examples of basal docking and how apico-basolateral polarity changes during epithelial fusion. Understanding the specific molecular mechanisms of basal docking is an area ripe for further exploration that will shed light on complex morphogenetic events that sculpt developing organisms and on the cellular mechanisms that can go awry during diseases involving the formation of cysts, fistulas, atresias, and metastases.
Collapse
|
14
|
Altered cytoskeletal status in the transition from proneural to mesenchymal glioblastoma subtypes. Sci Rep 2022; 12:9838. [PMID: 35701472 PMCID: PMC9197936 DOI: 10.1038/s41598-022-14063-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 05/31/2022] [Indexed: 11/26/2022] Open
Abstract
Glioblastoma is a highly aggressive brain tumor with poor patient prognosis. Treatment outcomes remain limited, partly due to intratumoral heterogeneity and the invasive nature of the tumors. Glioblastoma cells invade and spread into the surrounding brain tissue, and even between hemispheres, thus hampering complete surgical resection. This invasive motility can arise through altered properties of the cytoskeleton. We hypothesize that cytoskeletal organization and dynamics can provide important clues to the different malignant states of glioblastoma. In this study, we investigated cytoskeletal organization in glioblastoma cells with different subtype expression profiles, and cytoskeletal dynamics upon subtype transitions. Analysis of the morphological, migratory, and invasive properties of glioblastoma cells identified cytoskeletal components as phenotypic markers that can serve as diagnostic or prognostic tools. We also show that the cytoskeletal function and malignant properties of glioblastoma cells shift during subtype transitions induced by altered expression of the neurodevelopmental transcription factor SOX2. The potential of SOX2 re-expression to reverse the mesenchymal subtype into a more proneural subtype might open up strategies for novel glioblastoma treatments.
Collapse
|
15
|
Aly NAR, Rizk S, Aboul Enein A, El Desoukey N, Zawam H, Ahmed M, El Shikh ME, Pitzalis C. The role of lymphoid tissue SPARC in the pathogenesis and response to treatment of multiple myeloma. Front Oncol 2022; 12:1009993. [PMID: 36605435 PMCID: PMC9807864 DOI: 10.3389/fonc.2022.1009993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Background Despite the significant progress in the treatment of multiple myeloma (MM), the disease remains untreatable and its cure is still an unmet clinical need. Neoplastic transformation in MM is initiated in the germinal centers (GCs) of secondary lymphoid tissue (SLT) where B cells experience extensive somatic hypermutation induced by follicular dendritic cells (FDCs) and T-cell signals. Objective We reason that secreted protein acidic and rich in cysteine (SPARC), a common stromal motif expressed by FDCs at the origin (SLTs) and the destination (BM) of MM, plays a role in the pathogenesis of MM, and, here, we sought to investigate this role. Methods There were 107 BM biopsies from 57 MM patients (taken at different time points) together with 13 control specimens assessed for SPARC gene and protein expression and compared with tonsillar tissues. In addition, regulation of myeloma-promoting genes by SPARC-secreting FDCs was assessed in in vitro GC reactions (GCRs). Results SPARC gene expression was confirmed in both human primary (BM) and secondary (tonsils) lymphoid tissues, and the expression was significantly higher in the BM. Sparc was detectable in the BM and tonsillar lysates, co-localized with the FDC markers in both tissues, and stimulation of FDCs in vitro induced significantly higher levels of SPARC expression than unstimulated controls. In addition, SPARC inversely correlated with BM PC infiltration, ISS staging, and ECOG performance of the MM patients, and in vitro addition of FDCs to lymphocytes inhibited the expression of several oncogenes associated with malignant transformation of PCs. Conclusion FDC-SPARC inhibits several myelomagenic gene expression and inversely correlates with PC infiltration and MM progression. Therapeutic induction of SPARC expression through combinations of the current MM drugs, repositioning of non-MM drugs, or novel drug discovery could pave the way to better control MM in clinically severe and drug-resistant patients.
Collapse
Affiliation(s)
- Nesreen Amer Ramadan Aly
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Samia Rizk
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Azza Aboul Enein
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nermeen El Desoukey
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hamdy Zawam
- Clinical Oncology and Nuclear Radiation Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Manzoor Ahmed
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mohey Eldin El Shikh
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- *Correspondence: Mohey Eldin El Shikh,
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
16
|
Smith JJ, Xiao Y, Parsan N, Medwig-Kinney TN, Martinez MAQ, Moore FEQ, Palmisano NJ, Kohrman AQ, Chandhok Delos Reyes M, Adikes RC, Liu S, Bracht SA, Zhang W, Wen K, Kratsios P, Matus DQ. The SWI/SNF chromatin remodeling assemblies BAF and PBAF differentially regulate cell cycle exit and cellular invasion in vivo. PLoS Genet 2022; 18:e1009981. [PMID: 34982771 PMCID: PMC8759636 DOI: 10.1371/journal.pgen.1009981] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/14/2022] [Accepted: 12/07/2021] [Indexed: 12/15/2022] Open
Abstract
Chromatin remodelers such as the SWI/SNF complex coordinate metazoan development through broad regulation of chromatin accessibility and transcription, ensuring normal cell cycle control and cellular differentiation in a lineage-specific and temporally restricted manner. Mutations in genes encoding the structural subunits of chromatin, such as histone subunits, and chromatin regulating factors are associated with a variety of disease mechanisms including cancer metastasis, in which cancer co-opts cellular invasion programs functioning in healthy cells during development. Here we utilize Caenorhabditis elegans anchor cell (AC) invasion as an in vivo model to identify the suite of chromatin agents and chromatin regulating factors that promote cellular invasiveness. We demonstrate that the SWI/SNF ATP-dependent chromatin remodeling complex is a critical regulator of AC invasion, with pleiotropic effects on both G0 cell cycle arrest and activation of invasive machinery. Using targeted protein degradation and enhanced RNA interference (RNAi) vectors, we show that SWI/SNF contributes to AC invasion in a dose-dependent fashion, with lower levels of activity in the AC corresponding to aberrant cell cycle entry and increased loss of invasion. Our data specifically implicate the SWI/SNF BAF assembly in the regulation of the G0 cell cycle arrest in the AC, whereas the SWI/SNF PBAF assembly promotes AC invasion via cell cycle-independent mechanisms, including attachment to the basement membrane (BM) and activation of the pro-invasive fos-1/FOS gene. Together these findings demonstrate that the SWI/SNF complex is necessary for two essential components of AC invasion: arresting cell cycle progression and remodeling the BM. The work here provides valuable single-cell mechanistic insight into how the SWI/SNF assemblies differentially contribute to cellular invasion and how SWI/SNF subunit-specific disruptions may contribute to tumorigeneses and cancer metastasis. Cellular invasion is required for animal development and homeostasis. Inappropriate activation of invasion however can result in cancer metastasis. Invasion programs are orchestrated by complex gene regulatory networks (GRN) that function in a coordinated fashion to turn on and off pro-invasive genes. While the core of GRNs are DNA binding transcription factors, they require aid from chromatin remodelers to access the genome. To identify the suite of pro-invasive chromatin remodelers, we paired high resolution imaging with RNA interference to individually knockdown 269 chromatin factors, identifying the evolutionarily conserved SWItching defective/Sucrose Non-Fermenting (SWI/SNF) ATP-dependent chromatin remodeling complex as a new regulator of Caenorhabditis elegans anchor cell (AC) invasion. Using a combination of CRISPR/Cas9 genome engineering and targeted protein degradation we demonstrate that the core SWI/SNF complex functions in a dose-dependent manner to control invasion. Further, we determine that the accessory SWI/SNF complexes, BAF and PBAF, contribute to invasion via distinctive mechanisms: BAF is required to prevent inappropriate proliferation while PBAF promotes AC attachment and remodeling of the basement membrane. Together, our data provide insights into how the SWI/SNF complex, which is mutated in many human cancers, can function in a dose-dependent fashion to regulate switching from invasive to proliferative fates.
Collapse
Affiliation(s)
- Jayson J. Smith
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Yutong Xiao
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Nithin Parsan
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Taylor N. Medwig-Kinney
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Michael A. Q. Martinez
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Frances E. Q. Moore
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Nicholas J. Palmisano
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Abraham Q. Kohrman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Mana Chandhok Delos Reyes
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Rebecca C. Adikes
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Biology Department, Siena College, Loudonville, New York, United States of America
| | - Simeiyun Liu
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Molecular, Cellular and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Sydney A. Bracht
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Department of Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Wan Zhang
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Kailong Wen
- The Grossman Institute for Neuroscience, Quantitative Biology, and Human Behavior, University of Chicago, Chicago, Illinois, United States of America
- Department of Neurobiology, University of Chicago, Chicago, Illinois, United States of America
| | - Paschalis Kratsios
- The Grossman Institute for Neuroscience, Quantitative Biology, and Human Behavior, University of Chicago, Chicago, Illinois, United States of America
- Department of Neurobiology, University of Chicago, Chicago, Illinois, United States of America
| | - David Q. Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
17
|
Zhang S, Xiang X, Liu L, Yang H, Cen D, Tang G. Bioinformatics Analysis of Hub Genes and Potential Therapeutic Agents Associated with Gastric Cancer. Cancer Manag Res 2021; 13:8929-8951. [PMID: 34876855 PMCID: PMC8643151 DOI: 10.2147/cmar.s341485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/16/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose The current treatment methods available for advanced gastric cancer are not very promising. Hence, it is important to explore novel biomarkers and potential therapeutic agents to treat gastric cancer (GC). This study aimed to identify hub genes associated with GC prognosis and explore potential drugs for its treatment. Materials and Methods Three gene expression data of GC and normal tissues were downloaded from the Gene Expression Omnibus (GEO) and processed to identify the differentially expressed genes (DEGs). We conducted a comprehensive analysis of DEGs, including functional enrichment analysis, construction of protein-protein interaction (PPI) network, identification of hub genes, survival analysis and expression verification of hub genes. Finally, we constructed the network of miRNA-mRNA, and predicted the drugs that might be effective for GC treatment. Results A total of 340 DEGs, including 94 up-regulated and 246 down-regulated genes, were identified. Among the up-regulated DEGs, the enrichment terms were primarily related to tumorigenesis and tumor progression, extracellular matrix organization, and collagen catabolic process. Additionally, 10 hub genes (FN1, COL3A1, COL1A2, BGN, THBS2, COL5A2, THBS1, COL5A1, SPARC, and COL4A1) were identified, out of which 7 genes were significantly associated with poor overall survival (OS) in GC. The expression levels of these 7 hub genes were verified using real-time PCR, immunohistochemistry, and the GEPIA2 (Gene Expression Profiling Interactive Analysis) server. A regulatory network of miRNA-mRNA was also constructed, and the top 4 interactive miRNAs (hsa-miR-29b-3p, hsa-miR-140-3p, hsa-miR-29a-3p, and hsa-miR-29c-3p) that targeted the most hub genes were identified. Finally, fourteen small molecules were predicted to be effective in treating GC. Conclusion The identification of the hub genes, miRNA-mRNA network, and potential candidate drugs associated with GC provides new insights into the molecular mechanisms and treatment of GC.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning City, Guangxi Province, People's Republic of China
| | - Xuelian Xiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning City, Guangxi Province, People's Republic of China
| | - Li Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning City, Guangxi Province, People's Republic of China
| | - Huiying Yang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning City, Guangxi Province, People's Republic of China
| | - Dongliang Cen
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning City, Guangxi Province, People's Republic of China
| | - Guodu Tang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning City, Guangxi Province, People's Republic of China
| |
Collapse
|
18
|
Zhu YS, Zhang JN, Mo TT, Jiang C, Ma RC, Chen L. Discoidin domain receptor 2 activation of p38 mitogen-activated protein kinase as an important pathway for osteonectin-regulating osteoblast mineralization. J Orthop Surg Res 2021; 16:711. [PMID: 34876214 PMCID: PMC8650413 DOI: 10.1186/s13018-021-02860-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022] Open
Abstract
Objective The present study aimed to determine the role of the discoidin domain receptor 2 (DDR2) in the osteonectin (ON) regulation of osteoblast mineralization through the activation of p38 mitogen-activated protein kinase (MAPK). Methods Four groups were established: the ON group, the inhibitor group, the Ddr2-small interfering ribonucleic acid (siRNA) group, and the control group. Osteoblasts from the parietal bones of neonatal Sprague–Dawley rats were isolated and cultured. In the ON group, 1 µg/mL ON was added to the osteoblasts. The gene expressions of collagen 1 (Col 1) and Ddr2 were detected using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). In the inhibitor group, the osteoblasts were added to WRG-28 (a specific DDR2 inhibitor), and in the Ddr2-siRNA group, the osteoblasts were transfected with Ddr2-siRNA. The gene and protein expressions of DDR2, bone sialoprotein, osteocalcin, osteopontin, and p38 MAPK were determined using RT-qPCR and western blot analysis. Alizarin red staining and transmission electron microscopy were used to detect mineralization. Results The results showed that ON enhanced the osteoblast Col 1 and Ddr2 gene expressions, while the use of a Ddr2-siRNA/DDR2-blocker decreased the OPN, BSP, OCN, and P38 gene and protein expressions and reduced osteoblast cellular activity and mineralized nodules. Conclusion The present study demonstrated that DDR2 activation of p38 MAPK is an important approach to ON-regulating osteoblast mineralization.
Collapse
Affiliation(s)
- Yun-Sen Zhu
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China
| | - Jiang-Nan Zhang
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China
| | - Ting-Ting Mo
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China
| | - Chang Jiang
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China.
| | - Ru-Chao Ma
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China
| | - Liang Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China
| |
Collapse
|
19
|
Costa AC, Santos JMO, Gil da Costa RM, Medeiros R. Impact of immune cells on the hallmarks of cancer: A literature review. Crit Rev Oncol Hematol 2021; 168:103541. [PMID: 34801696 DOI: 10.1016/j.critrevonc.2021.103541] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/15/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022] Open
Abstract
Tumor-infiltrating immune cells (TIICs) are critical players in the tumor microenvironment, modulating cancer cell functions. TIICs are highly heterogenic and plastic and may either suppress cancers or provide support for tumor growth. A wide range of studies have shed light on how tumor-associated macrophages, dendritic cells, neutrophils, mast cells, natural killer cells and lymphocytes contribute for the establishment of several hallmarks of cancer and became the basis for successful immunotherapies. Many of those TIICs play pivotal roles in several hallmarks of cancer. This review contributes to elucidate the multifaceted roles of immune cells in cancer development, highlighting molecular components that constitute promising therapeutic targets. Additional studies are needed to clarify the relation between TIICs and hallmarks such as enabling replicative immortality, evading growth suppressors, sustaining proliferative signaling, resisting cell death and genome instability and mutation, to further explore their therapeutic potential and improve the outcomes of cancer patients.
Collapse
Affiliation(s)
- Alexandra C Costa
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Medicine of the University of Porto (FMUP), 4200-319, Porto, Portugal.
| | - Joana M O Santos
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Medicine of the University of Porto (FMUP), 4200-319, Porto, Portugal.
| | - Rui M Gil da Costa
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal; LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal; Postgraduate Programme in Adult Health (PPGSAD), Department of Morphology, Federal University of Maranhão (UFMA), and UFMA University Hospital (HUUFMA), 65080-805, São Luís, Brazil.
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Medicine of the University of Porto (FMUP), 4200-319, Porto, Portugal; Virology Service, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072, Porto, Portugal; CEBIMED, Faculty of Health Sciences of the Fernando Pessoa University, 4249-004, Porto, Portugal; Research Department of the Portuguese League Against Cancer-Regional Nucleus of the North (Liga Portuguesa Contra o Cancro-Núcleo Regional do Norte), 4200-177, Porto, Portugal.
| |
Collapse
|
20
|
Garde A, Sherwood DR. Fueling Cell Invasion through Extracellular Matrix. Trends Cell Biol 2021; 31:445-456. [PMID: 33549396 PMCID: PMC8122022 DOI: 10.1016/j.tcb.2021.01.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 01/10/2023]
Abstract
Cell invasion through extracellular matrix (ECM) has pivotal roles in cell dispersal during development, immune cell trafficking, and cancer metastasis. Many elegant studies have revealed the specialized cellular protrusions, proteases, and distinct modes of migration invasive cells use to overcome ECM barriers. Less clear, however, is how invasive cells provide energy, specifically ATP, to power the energetically demanding membrane trafficking, F-actin polymerization, and actomyosin machinery that mediate break down, remodeling, and movement through ECMs. Here, we provide an overview of the challenges of examining ATP generation and delivery within invading cells and how recent studies using diverse invasion models, experimental approaches, and energy biosensors are revealing that energy metabolism is an integral component of cell invasive behavior that is dynamically tuned to overcome the ECM environment.
Collapse
Affiliation(s)
- Aastha Garde
- Department of Cell Biology, Duke University, Box 3709, Durham, NC 27710, USA
| | - David R Sherwood
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA; Regeneration Next, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
21
|
Yang X, Wu Q, Wu F, Zhong Y. Differential expression of COL4A3 and collagen in upward and downward progressing types of nasopharyngeal carcinoma. Oncol Lett 2021; 21:223. [PMID: 33613712 PMCID: PMC7859474 DOI: 10.3892/ol.2021.12484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022] Open
Abstract
Upward (local growth and invasion of the base of skull), downward (distant metastasis) and mixed progressing types of nasopharyngeal carcinoma (NPC) have been identified and are distinctly different with respect to clinical symptoms, therapeutic strategies and prognosis. The present study aimed to identify the genetic difference and collagen expression levels in the upward and downward progressing types of NPC. Whole exon sequencing (WES) was used to detect genes differentially mutated between the upward and downward progressing types of NPC. Collagen deposition in the upward and downward progressing types of NPC was determined using Masson trichromatic staining, while the protein expression level of COL4A3 was detected using immunohistochemistry. Survival analysis was also performed using the Kaplan-Meier Plotter database to examine the role of COL4A3 expression level in the prognosis of head and neck squamous cell carcinoma. Knockdown of COL4A3 was performed using short interfering (si)RNA-COL4A3 in a 5-8F NPC cell line. Reverse transcription-quantitative PCR and western blot analyses were utilized to analyze the mRNA and protein expression levels of COL4A3, respectively. The roles of COL4A3 in the migration and invasion of the 5-8F cell line were examined using wound-healing Transwell and Matrigel assays, respectively. A total of 21 genes were differentially mutated between the upward and downward progressing types of NPC. The COL4A3 was investigated further, as it was found to be associated with extracellular matrix deposition and cancer metastasis. The COL4A3 gene was markedly downregulated in the downward progressing type compared with that in the upward progressing type (2.161±1.306 vs. 5.077±3.619; P<0.05). In addition, the deposition of collagen in the downward progressing type was also significantly decreased compared with that in the upward progressing type (5.63±6.83 vs. 10.94±9.60; P<0.05). Kaplan-Meier analysis indicated that high expression level of COL4A3 was positively associated with a favorable prognosis of head and neck squamous cell carcinoma (HR, 0.69; 95% CI, 0.49- 0.97; P=0.031). To confirm the role of COL4A3, the expression level of COL4A3 was knocked down using siRNA in the 5-8F cell line and the results showed that the invasion and migration was significantly increased when the expression of COL4A3 was inhibited (P<0.0001). In conclusion, the gene mutation patterns were significantly different between the upward and downward progressing types of NPC. In addition, the expression level of the COL4A3 gene was decreased in the downward progressing type, which might promote NPC metastasis through the downregulation of extracellular collagen expression.
Collapse
Affiliation(s)
- Xiting Yang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Fengyang Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yahua Zhong
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
- Correspondence to: Professor Yahua Zhong, Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang, Wuhan, Hubei 430071, P.R. China, E-mail:
| |
Collapse
|
22
|
McGovern KE, Nance JP, David CN, Harrison RES, Noor S, Worth D, Landrith TA, Obenaus A, Carson MJ, Morikis D, Wilson EH. SPARC coordinates extracellular matrix remodeling and efficient recruitment to and migration of antigen-specific T cells in the brain following infection. Sci Rep 2021; 11:4549. [PMID: 33633185 PMCID: PMC7907143 DOI: 10.1038/s41598-021-83952-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/03/2021] [Indexed: 01/14/2023] Open
Abstract
Central nervous system (CNS) injury and infection can result in profound tissue remodeling in the brain, the mechanism and purpose of which is poorly understood. Infection with the protozoan parasite Toxoplasma gondii causes chronic infection and inflammation in the brain parenchyma. Control of parasite replication requires the continuous presence of IFNγ-producing T cells to keep T. gondii in its slowly replicating cyst form. During infection, a network of extracellular matrix fibers, revealed using multiphoton microscopy, forms in the brain. The origin and composition of these structures are unknown but the fibers have been observed to act as a substrate for migrating T cells. In this study, we show a critical regulator of extracellular matrix (ECM) remodeling, Secreted Protein, Acidic, Rich in Cysteine (SPARC), is upregulated in the brain during the early phases of infection in the frontal cortex. In the absence of SPARC, a reduced and disordered fibrous network, increased parasite burden, and reduced antigen-specific T cell entry into the brain points to a role for SPARC in T cell recruitment to and migration within the brain. We also report SPARC can directly bind to CCR7 ligands CCL19 and CCL21 but not CXCL10, and enhance migration toward a chemokine gradient. Measurement of T cell behavior points to tissue remodeling being important for access of immune cells to the brain and facilitating cellular locomotion. Together, these data identify SPARC as an important regulatory component of immune cell trafficking and access to the inflamed CNS.
Collapse
Affiliation(s)
- Kathryn E McGovern
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- BIO5 Institute, Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
| | - J Philip Nance
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Clément N David
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- Nanostring Technologies, Inc, 530 Fairview Ave N, Seattle, WA, 98109, USA
| | - Reed E S Harrison
- Department of Bioengineering, University of California, Riverside, Riverside, CA, 92521-0129, USA
- UCSD Bioengineering and the Institute for Engineering in Medicine, San Diego, CA, 92093, USA
| | - Shahani Noor
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- School of Medicine, MSC08, University of New Mexico, Albequerque, NM, 87131, USA
| | - Danielle Worth
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Tyler A Landrith
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- Ambrey Genetics, Aliso Viejo, CA, 92656, USA
| | - Andre Obenaus
- School of Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Monica J Carson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Dimitrios Morikis
- Department of Bioengineering, University of California, Riverside, Riverside, CA, 92521-0129, USA
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
23
|
Martins Cavaco AC, Dâmaso S, Casimiro S, Costa L. Collagen biology making inroads into prognosis and treatment of cancer progression and metastasis. Cancer Metastasis Rev 2021; 39:603-623. [PMID: 32447477 DOI: 10.1007/s10555-020-09888-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progression through dissemination to tumor-surrounding tissues and metastasis development is a hallmark of cancer that requires continuous cell-to-cell interactions and tissue remodeling. In fact, metastization can be regarded as a tissue disease orchestrated by cancer cells, leading to neoplastic colonization of new organs. Collagen is a major component of the extracellular matrix (ECM), and increasing evidence suggests that it has an important role in cancer progression and metastasis. Desmoplasia and collagen biomarkers have been associated with relapse and death in cancer patients. Despite the increasing interest in ECM and in the desmoplastic process in tumor microenvironment as prognostic factors and therapeutic targets in cancer, further research is required for a better understanding of these aspects of cancer biology. In this review, published evidence correlating collagen with cancer prognosis is retrieved and analyzed, and the role of collagen and its fragments in cancer pathophysiology is discussed.
Collapse
Affiliation(s)
- Ana C Martins Cavaco
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Sara Dâmaso
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, 1649-028, Lisboa, Portugal
| | - Sandra Casimiro
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Luís Costa
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal.
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, 1649-028, Lisboa, Portugal.
| |
Collapse
|
24
|
The extracellular matrix: A key player in the pathogenesis of hematologic malignancies. Blood Rev 2020; 48:100787. [PMID: 33317863 DOI: 10.1016/j.blre.2020.100787] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/10/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022]
Abstract
Hematopoietic stem and progenitor cells located in the bone marrow lay the foundation for multiple lineages of mature hematologic cells. Bone marrow niches are architecturally complex with specific cellular, physiochemical, and biomechanical factors. Increasing evidence suggests that the bone marrow microenvironment contributes to the pathogenesis of hematological neoplasms. Numerous studies have deciphered the role of genetic mutations and chromosomal translocations in the development hematologic malignancies. Significant progress has also been made in understanding how the cellular components and cytokine interactions within the bone marrow microenvironment promote the evolution of hematologic cancers. Although the extracellular matrix is known to be a key player in the pathogenesis of various diseases, it's role in the progression of hematologic malignancies is less understood. In this review, we discuss the interactions between the extracellular matrix and malignant cells, and provide an overview of the role of extracellular matrix remodeling in sustaining hematologic malignancies.
Collapse
|
25
|
Roig-Rosello E, Rousselle P. The Human Epidermal Basement Membrane: A Shaped and Cell Instructive Platform That Aging Slowly Alters. Biomolecules 2020; 10:E1607. [PMID: 33260936 PMCID: PMC7760980 DOI: 10.3390/biom10121607] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
One of the most important functions of skin is to act as a protective barrier. To fulfill this role, the structural integrity of the skin depends on the dermal-epidermal junction-a complex network of extracellular matrix macromolecules that connect the outer epidermal layer to the underlying dermis. This junction provides both a structural support to keratinocytes and a specific niche that mediates signals influencing their behavior. It displays a distinctive microarchitecture characterized by an undulating pattern, strengthening dermal-epidermal connectivity and crosstalk. The optimal stiffness arising from the overall molecular organization, together with characteristic anchoring complexes, keeps the dermis and epidermis layers extremely well connected and capable of proper epidermal renewal and regeneration. Due to intrinsic and extrinsic factors, a large number of structural and biological changes accompany skin aging. These changes progressively weaken the dermal-epidermal junction substructure and affect its functions, contributing to the gradual decline in overall skin physiology. Most changes involve reduced turnover or altered enzymatic or non-enzymatic post-translational modifications, compromising the mechanical properties of matrix components and cells. This review combines recent and older data on organization of the dermal-epidermal junction, its mechanical properties and role in mechanotransduction, its involvement in regeneration, and its fate during the aging process.
Collapse
Affiliation(s)
- Eva Roig-Rosello
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR 5305, CNRS-Université Lyon 1, SFR BioSciences Gerland-Lyon Sud, 7 Passage du Vercors, 69367 Lyon, France;
- Roger Gallet SAS, 4 rue Euler, 75008 Paris, France
| | - Patricia Rousselle
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR 5305, CNRS-Université Lyon 1, SFR BioSciences Gerland-Lyon Sud, 7 Passage du Vercors, 69367 Lyon, France;
| |
Collapse
|
26
|
Abstract
The glomerular basement membrane (GBM) is a key component of the glomerular capillary wall and is essential for kidney filtration. The major components of the GBM include laminins, type IV collagen, nidogens and heparan sulfate proteoglycans. In addition, the GBM harbours a number of other structural and regulatory components and provides a reservoir for growth factors. New technologies have improved our ability to study the composition and assembly of basement membranes. We now know that the GBM is a complex macromolecular structure that undergoes key transitions during glomerular development. Defects in GBM components are associated with a range of hereditary human diseases such as Alport syndrome, which is caused by defects in the genes COL4A3, COL4A4 and COL4A5, and Pierson syndrome, which is caused by variants in LAMB2. In addition, the GBM is affected by acquired autoimmune disorders and metabolic diseases such as diabetes mellitus. Current treatments for diseases associated with GBM involvement aim to reduce intraglomerular pressure and to treat the underlying cause where possible. As our understanding about the maintenance and turnover of the GBM improves, therapies to replace GBM components or to stimulate GBM repair could translate into new therapies for patients with GBM-associated disease.
Collapse
|
27
|
Keeley DP, Hastie E, Jayadev R, Kelley LC, Chi Q, Payne SG, Jeger JL, Hoffman BD, Sherwood DR. Comprehensive Endogenous Tagging of Basement Membrane Components Reveals Dynamic Movement within the Matrix Scaffolding. Dev Cell 2020; 54:60-74.e7. [PMID: 32585132 DOI: 10.1016/j.devcel.2020.05.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/09/2020] [Accepted: 05/15/2020] [Indexed: 12/18/2022]
Abstract
Basement membranes (BMs) are supramolecular matrices built on laminin and type IV collagen networks that provide structural and signaling support to tissues. BM complexity, however, has hindered an understanding of its formation, dynamics, and regulation. Using genome editing, we tagged 29 BM matrix components and receptors in C. elegans with mNeonGreen. Here, we report a common template that initiates BM formation, which rapidly diversifies during tissue differentiation. Through photobleaching studies, we show that BMs are not static-surprisingly, many matrix proteins move within the laminin and collagen scaffoldings. Finally, quantitative imaging, conditional knockdown, and optical highlighting indicate that papilin, a poorly studied glycoprotein, is the most abundant component in the gonadal BM, where it facilitates type IV collagen removal during BM expansion and tissue growth. Together, this work introduces methods for holistic investigation of BM regulation and reveals that BMs are highly dynamic and capable of rapid change to support tissues.
Collapse
Affiliation(s)
- Daniel P Keeley
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Eric Hastie
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Ranjay Jayadev
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Laura C Kelley
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Qiuyi Chi
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Sara G Payne
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA; Department of Cell Biology, Duke University, Box 3709, Durham, NC 27710, USA
| | - Jonathan L Jeger
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Box 90281, Durham, NC 27708, USA
| | - David R Sherwood
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA; Regeneration Next Initiative, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
28
|
Pastor-Pareja JC. Atypical basement membranes and basement membrane diversity - what is normal anyway? J Cell Sci 2020; 133:133/8/jcs241794. [PMID: 32317312 DOI: 10.1242/jcs.241794] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The evolution of basement membranes (BMs) played an essential role in the organization of animal cells into tissues and diversification of body plans. The archetypal BM is a compact extracellular matrix polymer containing laminin, nidogen, collagen IV and perlecan (LNCP matrix) tightly packed into a homogenously thin planar layer. Contrasting this clear-cut morphological and compositional definition, there are numerous examples of LNCP matrices with unusual characteristics that deviate from this planar organization. Furthermore, BM components are found in non-planar matrices that are difficult to categorize as BMs at all. In this Review, I discuss examples of atypical BM organization. First, I highlight atypical BM structures in human tissues before describing the functional dissection of a plethora of BMs and BM-related structures in their tissue contexts in the fruit fly Drosophila melanogaster To conclude, I summarize our incipient understanding of the mechanisms that provide morphological, compositional and functional diversity to BMs. It is becoming increasingly clear that atypical BMs are quite prevalent, and that even typical planar BMs harbor a lot of diversity that we do not yet comprehend.
Collapse
Affiliation(s)
- José C Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing 100084, China .,Peking-Tsinghua Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
29
|
Fan J, Ji T, Wang K, Huang J, Wang M, Manning L, Dong X, Shi Y, Zhang X, Shao Z, Colón-Ramos DA. A muscle-epidermis-glia signaling axis sustains synaptic specificity during allometric growth in Caenorhabditis elegans. eLife 2020; 9:55890. [PMID: 32255430 PMCID: PMC7164957 DOI: 10.7554/elife.55890] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/05/2020] [Indexed: 02/06/2023] Open
Abstract
Synaptic positions underlie precise circuit connectivity. Synaptic positions can be established during embryogenesis and sustained during growth. The mechanisms that sustain synaptic specificity during allometric growth are largely unknown. We performed forward genetic screens in C. elegans for regulators of this process and identified mig-17, a conserved ADAMTS metalloprotease. Proteomic mass spectrometry, cell biological and genetic studies demonstrate that MIG-17 is secreted from cells like muscles to regulate basement membrane proteins. In the nematode brain, the basement membrane does not directly contact synapses. Instead, muscle-derived basement membrane coats one side of the glia, while glia contact synapses on their other side. MIG-17 modifies the muscle-derived basement membrane to modulate epidermal-glial crosstalk and sustain glia location and morphology during growth. Glia position in turn sustains the synaptic pattern established during embryogenesis. Our findings uncover a muscle-epidermis-glia signaling axis that sustains synaptic specificity during the organism's allometric growth.
Collapse
Affiliation(s)
- Jiale Fan
- Department of Neurosurgery, the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, the Institutes of Brain Science, and Zhongshan Hospital, Fudan University Shanghai, Shanghai, China
| | - Tingting Ji
- Department of Neurosurgery, the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, the Institutes of Brain Science, and Zhongshan Hospital, Fudan University Shanghai, Shanghai, China
| | - Kai Wang
- Department of Neurosurgery, the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, the Institutes of Brain Science, and Zhongshan Hospital, Fudan University Shanghai, Shanghai, China
| | - Jichang Huang
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Shanghai, China
| | - Mengqing Wang
- Department of Neurosurgery, the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, the Institutes of Brain Science, and Zhongshan Hospital, Fudan University Shanghai, Shanghai, China
| | - Laura Manning
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Xiaohua Dong
- Department of Neurosurgery, the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, the Institutes of Brain Science, and Zhongshan Hospital, Fudan University Shanghai, Shanghai, China
| | - Yanjun Shi
- Department of Neurosurgery, the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, the Institutes of Brain Science, and Zhongshan Hospital, Fudan University Shanghai, Shanghai, China
| | - Xumin Zhang
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhiyong Shao
- Department of Neurosurgery, the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, the Institutes of Brain Science, and Zhongshan Hospital, Fudan University Shanghai, Shanghai, China
| | - Daniel A Colón-Ramos
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, United States.,Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
30
|
Duncan S, Delage S, Chioran A, Sirbu O, Brown TJ, Ringuette MJ. The predicted collagen-binding domains of Drosophila SPARC are essential for survival and for collagen IV distribution and assembly into basement membranes. Dev Biol 2020; 461:197-209. [PMID: 32087195 DOI: 10.1016/j.ydbio.2020.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/29/2020] [Accepted: 02/15/2020] [Indexed: 11/17/2022]
Abstract
The assembly of basement membranes (BMs) into tissue-specific morphoregulatory structures requires non-core BM components. Work in Drosophila indicates a principal role of collagen-binding matricellular glycoprotein SPARC (Secreted Protein, Acidic, Rich in Cysteine) in larval fat body BM assembly. We report that SPARC and collagen IV (Col(IV)) first colocalize in the trans-Golgi of hemocyte-like cell lines. Mutating the collagen-binding domains of Drosophila SPARC led to the loss of colocalization with Col(IV), a fibrotic-like BM, and 2nd instar larval lethality, indicating that SPARC binding to Col(IV) is essential for survival. Analysis of this mutant at 2nd instar reveals increased Col(IV) puncta within adipocytes, reflecting a disruption in the intracellular chaperone-like activity of SPARC. Removal of the disulfide bridge in the C-terminal EF-hand2 of SPARC, which is known to enhance Col(IV) binding, did not lead to larval lethality; however, a less intense fat body phenotype was observed. Additionally, both SPARC mutants exhibited altered fat body BM pore topography. Wing imaginal disc-derived SPARC did not localize within Col(IV)-rich matrices. This raises the possibility that SPARC interaction with Col(IV) requires initial intracellular interaction to colocalize at the BM or that wing-derived SPARC undergoes differential post-translational modifications that impacts its function. Collectively, these data provide evidence that the chaperone-like activity of SPARC on Col(IV) begins just prior to their co-secretion and demonstrate for the first time that the Col(IV) chaperone-like activity of SPARC is necessary for Drosophila development beyond the 2nd instar.
Collapse
Affiliation(s)
- Sebastian Duncan
- Department of Cells and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Samuel Delage
- Department of Cells and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Alexa Chioran
- Department of Cells and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Olga Sirbu
- Department of Cells and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Theodore J Brown
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum, Research Institute at Sinai Health Systems, Toronto, Ontario, Canada
| | - Maurice J Ringuette
- Department of Cells and Systems Biology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
31
|
Ma Y, Chen H, Ma H, Yao Z, Hu J, Ma J, Zhang X, Chen G, Liu Y. Prognostic role of secreted protein acidic and rich in cysteine in patients with solid tumors. Saudi Med J 2020; 40:755-765. [PMID: 31423511 PMCID: PMC6718847 DOI: 10.15537/smj.2019.8.24379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES To analyze the heterogeneous functions of secreted protein acidic and rich in cysteine (SPARC) from different origins and in different tumor microenvironments with the purpose of determining its clinical significance. Methods: The PubMed, CINAHL, Cochrane, Web of Science and Embase databases were utilized. Studies that focused on the effects of SPARC expression on solid tumor progression and clinical implications were used. The different outcomes including overall survival and disease-free survival were analyzed to evaluate their relations with tumor- and stroma-derived SPARC expression. Results: A total of 26 studies including 5,939 patients were enrolled in the present meta-analysis. Tumor-derived SPARC overexpression was significantly related with poor overall survival (hazard ratio: 1.478; 95% CI: 1.143-1.910; p=0.003), and a similar tendency was also observed in disease-free survival (hazard ratio: 1.476; 95% CI: 0.993-2.195; p=0.054). However, the hazard ratios for overall survival and disease-free survival did not present a statistical trend in stromal SPARC overexpression. Tumor type subgroup analysis revealed marked heterogeneity among outcomes. In pancreatic cancer, SPARC overexpression in the stroma was significantly associated with poorer overall survival and disease-free survival. In colorectal cancer, SPARC overexpression in the stroma was associated with better disease-free survival. Conclusion: For the majority of solid tumors, SPARC in cancer cells may be an unfavorable indicator for long-term survival for patients. As for stromal expression, SPARC indicates a poorer prognosis in pancreatic cancer, but a better disease-free survival in colorectal cancer. Secreted protein acidic and rich in cysteine might be a potential biomarker for solid tumor prognosis.
Collapse
Affiliation(s)
- Yongchen Ma
- Department of General Surgery, Peking University First Hospital, Beijing, People's Republic of China. E-mail.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Baldari S, Di Rocco G, Toietta G. Current Biomedical Use of Copper Chelation Therapy. Int J Mol Sci 2020; 21:1069. [PMID: 32041110 PMCID: PMC7037088 DOI: 10.3390/ijms21031069] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Abstract
Copper is an essential microelement that plays an important role in a wide variety of biological processes. Copper concentration has to be finely regulated, as any imbalance in its homeostasis can induce abnormalities. In particular, excess copper plays an important role in the etiopathogenesis of the genetic disease Wilson's syndrome, in neurological and neurodegenerative pathologies such as Alzheimer's and Parkinson's diseases, in idiopathic pulmonary fibrosis, in diabetes, and in several forms of cancer. Copper chelating agents are among the most promising tools to keep copper concentration at physiological levels. In this review, we focus on the most relevant compounds experimentally and clinically evaluated for their ability to counteract copper homeostasis deregulation. In particular, we provide a general overview of the main disorders characterized by a pathological increase in copper levels, summarizing the principal copper chelating therapies adopted in clinical trials.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
- Department of Medical Surgical Sciences and Biotechnologies, University of Rome “La Sapienza”, C.so della Repubblica 79, 04100 Latina, Italy
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
| |
Collapse
|
33
|
Single-cell tracking demonstrates copper chaperone Atox1 to be required for breast cancer cell migration. Proc Natl Acad Sci U S A 2020; 117:2014-2019. [PMID: 31932435 PMCID: PMC6995000 DOI: 10.1073/pnas.1910722117] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Copper ions are needed for several hallmarks of cancer. However, the involved pathways, mechanisms, and copper-binding proteins are mostly unknown. We recently found that cytoplasmic Antioxidant 1 copper chaperone (Atox1), which is up-regulated in breast cancer, is localized at the lamellipodia edges of aggressive breast cancer cells. To reveal molecular insights into a putative role in cell migration, we here investigated breast cancer cell (MDA-MB-231) migration by video microscopy as a function of Atox1. Tracking of hundreds of individual cells (per condition) over a 9-h time series revealed that cell migration velocity and directionality are significantly reduced upon Atox1 silencing in the cells. Because silencing of the copper transporter ATP7A also reduced cell migration, these proteins appear to be on the same pathway, suggesting that their well-known copper transport activity is involved. In-cell proximity ligation assays demonstrated that Atox1, ATP7A, and the proenzyme of lysyl oxidase (LOX; copper-loaded via ATP7A) are all in close proximity and that LOX activity is reduced upon Atox1 silencing in the cells. Since LOX is an established player in cancer cell migration, our results imply that Atox1 mediates breast cancer cell migration via coordinated copper transport in the ATP7A-LOX axis. Because individual cell migration is an early step in breast cancer metastasis, Atox1 levels in tumor cells may be a predictive measure of metastasis potential and serve as a biomarker for copper depletion therapy.
Collapse
|
34
|
Ghanemi A, Melouane A, Yoshioka M, St-Amand J. Secreted protein acidic and rich in cysteine and bioenergetics: Extracellular matrix, adipocytes remodeling and skeletal muscle metabolism. Int J Biochem Cell Biol 2019; 117:105627. [PMID: 31589923 DOI: 10.1016/j.biocel.2019.105627] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022]
Abstract
The extracellular matrix (ECM) remodeling plays important roles in both adipocytes shape/expansion remodeling and the skeletal muscle (SM) metabolism. Secreted protein acidic and rich in cysteine (SPARC) is expressed in divers tissues including adipose tissue (AT) and SM where it impacts a variety of remodeling as well as metabolic functions. SPARC, also known as osteonectin or BM-40, is a glycoprotein associated with the ECM. Numerous researches attempted to elucidate the implications of SPARC in these two key metabolic tissues under different conditions. Whereas SPARC deficiency tends to shape the remodeling of the adipocytes and the fat distribution, this deficiency decreases SM metabolic properties. On the other hand, SPARC seems to be an enhancer of the metabolism and a mediator of the exercise-induced adaptation in the SM and as well as an adipogenesis inhibitor. Some findings about the SPARC effects on AT and SM seem "contradictory" in terms of tissue development and energy profile therefore highlighting the mechanistic role of SPARC in both is a priority. Yet, within this review, we expose selected researches and compare the results. We conclude with explanations to "reconcile" the different observations, hypothesize the feedback and regulatory character of SPARC and put its roles within the energetic and structural maps of both adipocytes and myocytes in homeostasis and in situations such as obesity or exercise. These properties explain the modifications and the remodeling seen in AT and SM undergoing adaptive changes (obesity, exercise, etc.) and represent a starting point for precise therapeutic targeting of SPARC-related pathways is conditions such as obesity, sarcopenia and diabetes.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Québec, Québec G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Québec G1V 0A6, Canada
| | - Aicha Melouane
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Québec, Québec G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Québec G1V 0A6, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Québec, Québec G1V 4G2, Canada
| | - Jonny St-Amand
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Québec, Québec G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Québec G1V 0A6, Canada.
| |
Collapse
|
35
|
Jayadev R, Chi Q, Keeley DP, Hastie EL, Kelley LC, Sherwood DR. α-Integrins dictate distinct modes of type IV collagen recruitment to basement membranes. J Cell Biol 2019; 218:3098-3116. [PMID: 31387941 PMCID: PMC6719451 DOI: 10.1083/jcb.201903124] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/16/2019] [Accepted: 07/10/2019] [Indexed: 01/04/2023] Open
Abstract
Basement membranes (BMs) are cell-associated extracellular matrices that support tissue integrity, signaling, and barrier properties. Type IV collagen is critical for BM function, yet how it is directed into BMs in vivo is unclear. Through live-cell imaging of endogenous localization, conditional knockdown, and misexpression experiments, we uncovered distinct mechanisms of integrin-mediated collagen recruitment to Caenorhabditis elegans postembryonic gonadal and pharyngeal BMs. The putative laminin-binding αINA-1/βPAT-3 integrin was selectively activated in the gonad and recruited laminin, which directed moderate collagen incorporation. In contrast, the putative Arg-Gly-Asp (RGD)-binding αPAT-2/βPAT-3 integrin was activated in the pharynx and recruited high levels of collagen in an apparently laminin-independent manner. Through an RNAi screen, we further identified the small GTPase RAP-3 (Rap1) as a pharyngeal-specific PAT-2/PAT-3 activator that modulates collagen levels. Together, these studies demonstrate that tissues can use distinct mechanisms to direct collagen incorporation into BMs to precisely control collagen levels and construct diverse BMs.
Collapse
Affiliation(s)
- Ranjay Jayadev
- Department of Biology, Regeneration Next, Duke University, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Qiuyi Chi
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Daniel P Keeley
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Eric L Hastie
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Laura C Kelley
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| |
Collapse
|
36
|
Riley HJ, Bradshaw AD. The Influence of the Extracellular Matrix in Inflammation: Findings from the SPARC-Null Mouse. Anat Rec (Hoboken) 2019; 303:1624-1629. [PMID: 30980479 DOI: 10.1002/ar.24133] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/26/2018] [Accepted: 10/11/2018] [Indexed: 01/08/2023]
Abstract
Matricellular proteins are secreted proteins that, among other functions, can contribute to extracellular matrix (ECM) assembly including modulation of cell:ECM interactions. Recent discoveries have indicated a fundamental role for the ECM in the regulation of inflammatory responses including cell extravasation and recruitment, immune cell differentiation, polarization, activation, and retention in tissues. Secreted protein acidic and rich in cysteine (SPARC) is a matricellular collagen-binding protein implicated in fibrillar collagen assembly in the ECM of connective tissue as well as in basal lamina organization. Functions of SPARC in modulating cell adhesion events are also reported. Studies of phenotypic responses observed in SPARC-null mice to a variety of injury models have yielded interesting insight into the functional importance of SPARC production and aberrations in ECM structure that occur in the absence of SPARC that influence immune cell behavior and inflammatory pathways. In this review, we will discuss several examples from different tissues in which SPARC-null mice exhibited an inflammatory response distinct from those of SPARC expressing mice and provide insight into novel ECM-dependent mechanisms that influence these responses. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hannah J Riley
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina
| | - Amy D Bradshaw
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Department of Veteran's Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
37
|
Pancreatic acinar differentiation is guided by differential laminin deposition. Sci Rep 2019; 9:2711. [PMID: 30804366 PMCID: PMC6389953 DOI: 10.1038/s41598-019-39077-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/12/2018] [Indexed: 01/03/2023] Open
Abstract
Endothelial cells play multiple roles during pancreas organogenesis. First, they are required to instruct endoderm-derived pancreatic progenitor cells to initiate branching morphogenesis. Later, blood vessels promote β-cell differentiation but also limit acinar development. In this work, we show how endothelial cells might signal to pancreatic progenitors and spatially regulate acinar differentiation. Using an ex vivo culture system of undifferentiated E12.5 pancreata, we demonstrate that embryonic endothelial progenitor cells and their conditioned medium prevent the expression of two members of the pro-acinar transcriptional PTF1L-complex. This effect is not mediated by SPARC, a protein abundantly released in the medium conditioned by endothelial progenitors. On the contrary, heterotrimeric laminin-α1β1γ1, also produced by endothelial progenitor cells, can repress acinar differentiation when used on its own on pancreatic explants. Lastly, we found that laminin-α1 is predominantly found in vivo around the pancreatic trunk cells, as compared to the tip cells, at E14.5. In conclusion, we propose that expression or deposition of laminin-α1β1γ1 around the trunk cells, where blood vessels are predominantly localized, prevent acinar differentiation of these cells. On the contrary, transient decreased expression or deposition of laminin-α1β1γ1 around the tip cells would allow PTF1L-complex formation and acinar differentiation.
Collapse
|
38
|
The in-silico characterization of the Caenorhabditis elegans matrisome and proposal of a novel collagen classification. Matrix Biol Plus 2019; 1:100001. [PMID: 33543001 PMCID: PMC7852208 DOI: 10.1016/j.mbplus.2018.11.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/07/2023] Open
Abstract
Proteins are the building blocks of life. While proteins and their localization within cells and sub-cellular compartments are well defined, the proteins predicted to be secreted to form the extracellular matrix - or matrisome - remain elusive in the model organism C. elegans. Here, we used a bioinformatic approach combining gene orthology and protein structure analysis and an extensive curation of the literature to define the C. elegans matrisome. Similar to the human genome, we found that 719 out of ~20,000 genes (~4%) of the C. elegans genome encodes matrisome proteins, including 181 collagens, 35 glycoproteins, 10 proteoglycans, and 493 matrisome-associated proteins. We report that 173 out of the 181 collagen genes are unique to nematodes and are predicted to encode cuticular collagens, which we are proposing to group into five clusters. To facilitate the use of our lists and classification by the scientific community, we developed an automated annotation tool to identify ECM components in large datasets. We also established a novel database of all C. elegans collagens (CeColDB). Last, we provide examples of how the newly defined C. elegans matrisome can be used for annotations and gene ontology analyses of transcriptomic, proteomic, and RNAi screening data. Because C. elegans is a widely used model organism for high throughput genetic and drug screens, and to study biological and pathological processes, the conserved matrisome genes may aid in identifying potential drug targets. In addition, the nematode-specific matrisome may be exploited for targeting parasitic infection of man and crops. Pipeline combining gene- and protein-sequence analysis to predict the C. elegans matrisome The in-silicoC. elegans matrisome comprises 719 genes. The 185 C. elegans collagen-domain-containing proteins are classified into 4 groups. The 173 cuticular collagens are further classified into 5 clusters based on their domain organization. The C. elegans Matrisome Annotator is an online tool to identify matrisome genes and proteins in large datasets.
Collapse
|
39
|
Keeley DP, Sherwood DR. Tissue linkage through adjoining basement membranes: The long and the short term of it. Matrix Biol 2019; 75-76:58-71. [PMID: 29803937 PMCID: PMC6252152 DOI: 10.1016/j.matbio.2018.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/18/2018] [Accepted: 05/21/2018] [Indexed: 01/01/2023]
Abstract
Basement membranes (BMs) are thin dense sheets of extracellular matrix that surround most tissues. When the BMs of neighboring tissues come into contact, they usually slide along one another and act to separate tissues and organs into distinct compartments. However, in certain specialized regions, the BMs of neighboring tissues link, helping to bring tissues together. These BM connections can be transient, such as during tissue fusion events in development, or long-term, as with adult tissues involved with filtration, including the blood brain barrier and kidney glomerulus. The transitory nature of these connections in development and the complexity of tissue filtration systems in adults have hindered the understanding of how juxtaposed BMs fasten together. The recent identification of a BM-BM adhesion system in C. elegans, termed B-LINK (BM linkage), however, is revealing cellular and extracellular matrix components of a nascent tissue adhesion system. We discuss insights gained from studying the B-LINK tissue adhesion system in C. elegans, compare this adhesion with other BM-BM connections in Drosophila and vertebrates, and outline important future directions towards elucidating this fascinating and poorly understood mode of adhesion that joins neighboring tissues.
Collapse
Affiliation(s)
- Daniel P Keeley
- Department of Biology, Regeneration Next, Duke University, Box 90338, Durham, NC 27708, USA
| | - David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Box 90338, Durham, NC 27708, USA.
| |
Collapse
|
40
|
Yan P, He Y, Xie K, Kong S, Zhao W. In silico analyses for potential key genes associated with gastric cancer. PeerJ 2018; 6:e6092. [PMID: 30568862 PMCID: PMC6287586 DOI: 10.7717/peerj.6092] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/09/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Understanding hub genes involved in gastric cancer (GC) metastasis could lead to effective approaches to diagnose and treat cancer. In this study, we aim to identify the hub genes and investigate the underlying molecular mechanisms of GC. METHODS To explore potential therapeutic targets for GC,three expression profiles (GSE54129, GSE33651, GSE81948) of the genes were extracted from the Gene Expression Omnibus (GEO) database. The GEO2R online tool was applied to screen out differentially expressed genes (DEGs) between GC and normal gastric samples. Database for Annotation, Visualization and Integrated Discovery was applied to perform Gene Ontology (GO) and KEGG pathway enrichment analysis. The protein-protein interaction (PPI) network of these DEGs was constructed using a STRING online software. The hub genes were identified by the CytoHubba plugin of Cytoscape software. Then, the prognostic value of these identified genes was verified by gastric cancer database derived from Kaplan-Meier plotter platform. RESULTS A total of 85 overlapped upregulated genes and 44 downregulated genes were identified. The majority of the DEGs were enriched in extracellular matrix organization, endodermal cell differentiation, and endoderm formation. Moreover, five KEGG pathways were significantly enriched, including ECM-receptor interaction, amoebiasis, AGE-RAGE signaling pathway in diabetic complications, focal adhesion, protein digestion and absorption. By combining the results of PPI network and CytoHubba, a total of nine hub genes including COL1A1, THBS1, MMP2, CXCL8, FN1, TIMP1, SPARC, COL4A1, and ITGA5 were selected. The Kaplan-Meier plotter database confirmed that overexpression levels of these genes were associated with reduced overall survival, except for THBS1 and CXCL8. CONCLUSIONS Our study suggests that COL1A1, MMP2, FN1, TIMP1, SPARC, COL4A1, and ITGA5 may be potential biomarkers and therapeutic targets for GC. Further study is needed to assess the effect of THBS1 and CXCL8 on GC.
Collapse
Affiliation(s)
- Ping Yan
- Department of Gastroenterology, Clinical College, Dali University, Dali, Yunnan, China
| | - Yingchun He
- Department of Clinical Laboratory, Clinical College, Dali University, Dali, Yunnan, China
| | - Kexin Xie
- Department of Clinical Laboratory, Clinical College, Dali University, Dali, Yunnan, China
| | - Shan Kong
- Department of Clinical Laboratory, Clinical College, Dali University, Dali, Yunnan, China
| | - Weidong Zhao
- Department of Clinical Laboratory, Clinical College, Dali University, Dali, Yunnan, China
| |
Collapse
|
41
|
Cáceres R, Bojanala N, Kelley LC, Dreier J, Manzi J, Di Federico F, Chi Q, Risler T, Testa I, Sherwood DR, Plastino J. Forces drive basement membrane invasion in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2018; 115:11537-11542. [PMID: 30348801 PMCID: PMC6233148 DOI: 10.1073/pnas.1808760115] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
During invasion, cells breach basement membrane (BM) barriers with actin-rich protrusions. It remains unclear, however, whether actin polymerization applies pushing forces to help break through BM, or whether actin filaments play a passive role as scaffolding for targeting invasive machinery. Here, using the developmental event of anchor cell (AC) invasion in Caenorhabditis elegans, we observe that the AC deforms the BM and underlying tissue just before invasion, exerting forces in the tens of nanonewtons range. Deformation is driven by actin polymerization nucleated by the Arp2/3 complex and its activators, whereas formins and cross-linkers are dispensable. Delays in invasion upon actin regulator loss are not caused by defects in AC polarity, trafficking, or secretion, as appropriate markers are correctly localized in the AC even when actin is reduced and invasion is disrupted. Overall force production emerges from this study as one of the main tools that invading cells use to promote BM disruption in C. elegans.
Collapse
Affiliation(s)
- Rodrigo Cáceres
- Laboratoire Physico-Chimie Curie, Institut Curie, PSL Research University, CNRS, 75005 Paris, France
- Université Paris Descartes, 75005 Paris, France
- Sorbonne Université, 75005 Paris, France
| | - Nagagireesh Bojanala
- Laboratoire Physico-Chimie Curie, Institut Curie, PSL Research University, CNRS, 75005 Paris, France
- Sorbonne Université, 75005 Paris, France
| | - Laura C Kelley
- Department of Biology, Regeneration Next, Duke University, Durham, NC 27705
| | - Jes Dreier
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 10014 Stockholm, Sweden
| | - John Manzi
- Laboratoire Physico-Chimie Curie, Institut Curie, PSL Research University, CNRS, 75005 Paris, France
- Sorbonne Université, 75005 Paris, France
| | - Fahima Di Federico
- Laboratoire Physico-Chimie Curie, Institut Curie, PSL Research University, CNRS, 75005 Paris, France
- Sorbonne Université, 75005 Paris, France
| | - Qiuyi Chi
- Department of Biology, Regeneration Next, Duke University, Durham, NC 27705
| | - Thomas Risler
- Laboratoire Physico-Chimie Curie, Institut Curie, PSL Research University, CNRS, 75005 Paris, France
- Sorbonne Université, 75005 Paris, France
| | - Ilaria Testa
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 10014 Stockholm, Sweden
| | - David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Durham, NC 27705
| | - Julie Plastino
- Laboratoire Physico-Chimie Curie, Institut Curie, PSL Research University, CNRS, 75005 Paris, France;
- Sorbonne Université, 75005 Paris, France
| |
Collapse
|
42
|
Sun W, Feng J, Yi Q, Xu X, Chen Y, Tang L. SPARC acts as a mediator of TGF-β1 in promoting epithelial-to-mesenchymal transition in A549 and H1299 lung cancer cells. Biofactors 2018; 44:453-464. [PMID: 30346081 DOI: 10.1002/biof.1442] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
Migration and metastasis of tumor cells greatly contributes to the failure of cancer treatment. Recently, the extracellular protein secreted protein acidic and rich in cysteine (SPARC) has been reported closely related to tumorigenesis. Some articles have suggested that SPARC promoted metastasis in several highly metastatic tumors. However, there are also some studies shown that SPARC acted as an antitumor factor. SPARC-induced epithelial-to-mesenchymal transition (EMT) in melanoma cells and promoted EMT in hepatocellular carcinoma. Therefore, the role of SPARC in tumorigenesis and its relationship with EMT is still unclear. In this study, we investigated the expression change of SPARC in A549 and H1299 lung cancer cells undergoing EMT process. Our study indicated that SPARC was upregulated in A549 and H1299 cells EMT process. We further investigated the function of SPARC on proliferation, migration, and EMT process of A549 and H1299 cells. Overexpression of SPARC promoted the migration and EMT of A549 and H1299 cells. Knockdown SPARC inhibited the EMT of A549 cells. Overexpression of SPARC induced the increased expression of p-Akt and P-ERK. Furthermore, exogenous SPARC peptide promoted transforming growth factor (TGF)-β1-induced EMT of A549 and H1299 cells. SPARC knockdown partially eliminated TGF-β1 function in inducing EMT of A549 cells. SPARC follistatin-like functional domain reduced the expression of E-cadherin, but had no effect on the expression of p-Akt and p-ERK. In conclusion, we elucidated that SPARC contributes to tumorigenesis by promoting migration and EMT of A549 and H1299 lung cancer cells. These results will provide some new suggestion for lung cancer treatment. © 2018 BioFactors, 44(5):453-464, 2018.
Collapse
Affiliation(s)
- Weichao Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Jianguo Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qian Yi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Department of Physiology, College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan Province, Luzhou, Sichuan Province, China
| | - Xichao Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ying Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
43
|
Muñoz D, Castillo H, Henríquez JP, Marcellini S. Bone regeneration after traumatic skull injury in Xenopus tropicalis. Mech Dev 2018; 154:153-161. [PMID: 30420272 DOI: 10.1016/j.mod.2018.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/29/2018] [Accepted: 06/30/2018] [Indexed: 10/28/2022]
Abstract
The main purpose of regenerative biology is to improve human health by exploiting cellular and molecular mechanisms favoring tissue repair. In recent years, non-mammalian vertebrates have emerged as powerful model organisms to tackle the problem of tissue regeneration. Here, we analyze the process of bone repair in metamorphosing Xenopus tropicalis tadpoles subjected to traumatic skull injury. Five days after skull perforation, a dense and highly vascularized mesenchymal is apparent over the injury site. Using an in vivo bone staining procedure based on independent pulses of Alizarin red and Calcein green, we show that the deposition of new bone matrix completely closes the wound in 15 days. The absence of cartilage implies that bone repair follows an intramembranous ossification route. Collagen second harmonic imaging reveals that while a well-organized lamellar type of bone is deposited during development, a woven type of bone is produced during the early-phase of the regeneration process. Osteoblasts lying against the regenerating bone robustly express fibrillar collagen 1a1, SPARC and Dlx5. These analyses establish Xenopus tropicalis as a new model system to improve traumatic skull injury recovery.
Collapse
Affiliation(s)
- David Muñoz
- Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Faculty of Biological Sciences, University of Concepción, Chile; Laboratory of Development and Evolution (LADE), University of Concepción, Chile
| | - Héctor Castillo
- Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Faculty of Biological Sciences, University of Concepción, Chile; Laboratory of Development and Evolution (LADE), University of Concepción, Chile
| | - Juan Pablo Henríquez
- Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Faculty of Biological Sciences, University of Concepción, Chile; Center for Advanced Microscopy (CMA Bio-Bio), University of Concepción, Chile
| | - Sylvain Marcellini
- Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Faculty of Biological Sciences, University of Concepción, Chile; Laboratory of Development and Evolution (LADE), University of Concepción, Chile.
| |
Collapse
|
44
|
Chen DY, Crest J, Bilder D. A Cell Migration Tracking Tool Supports Coupling of Tissue Rotation to Elongation. Cell Rep 2018; 21:559-569. [PMID: 29045826 DOI: 10.1016/j.celrep.2017.09.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 08/30/2017] [Accepted: 09/25/2017] [Indexed: 12/17/2022] Open
Abstract
Cell migration is indispensable to morphogenesis and homeostasis. Live imaging allows mechanistic insights, but long-term observation can alter normal biology, and tools to track movements in vivo without perturbation are lacking. We develop here a tool called M-TRAIL (matrix-labeling technique for real-time and inferred location), which reveals migration histories in fixed tissues. Using clones that overexpress GFP-tagged extracellular matrix (ECM) components, motility trajectories are mapped based on durable traces deposited onto basement membrane. We applied M-TRAIL to Drosophila follicle rotation, comparing in vivo and ex vivo migratory dynamics. The rate, trajectory, and cessation of rotation in wild-type (WT) follicles measured in vivo and ex vivo were identical, as was rotation failure in fat2 mutants. However, follicles carrying intracellularly truncated Fat2, previously reported to lack rotation ex vivo, in fact rotate in vivo at a reduced speed, thus revalidating the hypothesis that rotation is required for tissue elongation. The M-TRAIL approach could be applied to track and quantitate in vivo cell motility in other tissues and organisms.
Collapse
Affiliation(s)
- Dong-Yuan Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Justin Crest
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
45
|
Filipe EC, Chitty JL, Cox TR. Charting the unexplored extracellular matrix in cancer. Int J Exp Pathol 2018; 99:58-76. [PMID: 29671911 DOI: 10.1111/iep.12269] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is present in all solid tissues and considered a master regulator of cell behaviour and phenotype. The importance of maintaining the correct biochemical and biophysical properties of the ECM, and the subsequent regulation of cell and tissue homeostasis, is illustrated by the simple fact that the ECM is highly dysregulated in many different types of disease, especially cancer. The loss of tissue ECM homeostasis and integrity is seen as one of the hallmarks of cancer and typically defines transitional events in progression and metastasis. The vast majority of cancer studies place an emphasis on exploring the behaviour and intrinsic signalling pathways of tumour cells. Their goal was to identify ways to target intracellular pathways regulating cancer. Cancer progression and metastasis are powerfully influenced by the ECM and thus present a vast, unexplored repository of anticancer targets that we are only just beginning to tap into. Deconstructing the complexity of the tumour ECM landscape and identifying the interactions between the many cell types, soluble factors and extracellular-matrix proteins have proved challenging. Here, we discuss some of the emerging tools and platforms being used to catalogue and chart the ECM in cancer.
Collapse
Affiliation(s)
- Elysse C Filipe
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Jessica L Chitty
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Thomas R Cox
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
46
|
Abstract
The extracellular matrix (ECM) has central roles in tissue integrity and remodeling throughout the life span of animals. While collagens are the most abundant structural components of ECM in most tissues, tissue-specific molecular complexity is contributed by ECM glycoproteins. The matricellular glycoproteins are categorized primarily according to functional criteria and represented predominantly by the thrombospondin, tenascin, SPARC/osteonectin, and CCN families. These proteins do not self-assemble into ECM fibrils; nevertheless, they shape ECM properties through interactions with structural ECM proteins, growth factors, and cells. Matricellular proteins also promote cell migration or morphological changes through adhesion-modulating or counter-adhesive actions on cell-ECM adhesions, intracellular signaling, and the actin cytoskeleton. Typically, matricellular proteins are most highly expressed during embryonic development. In adult tissues, expression is more limited unless activated by cues for dynamic tissue remodeling and cell motility, such as occur during inflammatory response and wound repair. Many insights in the complex roles of matricellular proteins have been obtained from studies of gene knockout mice. However, with the exception of chordate-specific tenascins, these are highly conserved proteins that are encoded in many animal phyla. This review will consider the increasing body of research on matricellular proteins in nonmammalian animal models. These models provide better access to the very earliest stages of embryonic development and opportunities to study biological processes such as limb and organ regeneration. In aggregate, this research is expanding concepts of the functions and mechanisms of action of matricellular proteins.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
47
|
Matsubayashi Y, Louani A, Dragu A, Sánchez-Sánchez BJ, Serna-Morales E, Yolland L, Gyoergy A, Vizcay G, Fleck RA, Heddleston JM, Chew TL, Siekhaus DE, Stramer BM. A Moving Source of Matrix Components Is Essential for De Novo Basement Membrane Formation. Curr Biol 2017; 27:3526-3534.e4. [PMID: 29129537 PMCID: PMC5714436 DOI: 10.1016/j.cub.2017.10.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 08/11/2017] [Accepted: 10/02/2017] [Indexed: 12/31/2022]
Abstract
The basement membrane (BM) is a thin layer of extracellular matrix (ECM) beneath nearly all epithelial cell types that is critical for cellular and tissue function. It is composed of numerous components conserved among all bilaterians [1]; however, it is unknown how all of these components are generated and subsequently constructed to form a fully mature BM in the living animal. Although BM formation is thought to simply involve a process of self-assembly [2], this concept suffers from a number of logistical issues when considering its construction in vivo. First, incorporation of BM components appears to be hierarchical [3, 4, 5], yet it is unclear whether their production during embryogenesis must also be regulated in a temporal fashion. Second, many BM proteins are produced not only by the cells residing on the BM but also by surrounding cell types [6, 7, 8, 9], and it is unclear how large, possibly insoluble protein complexes [10] are delivered into the matrix. Here we exploit our ability to live image and genetically dissect de novo BM formation during Drosophila development. This reveals that there is a temporal hierarchy of BM protein production that is essential for proper component incorporation. Furthermore, we show that BM components require secretion by migrating macrophages (hemocytes) during their developmental dispersal, which is critical for embryogenesis. Indeed, hemocyte migration is essential to deliver a subset of ECM components evenly throughout the embryo. This reveals that de novo BM construction requires a combination of both production and distribution logistics allowing for the timely delivery of core components. Macrophages are major producers of basement membrane in the Drosophila embryo Basement membrane components require hierarchical deposition during development Macrophage migration is essential to evenly deliver a subset of matrix components Uneven macrophage dispersal leads to uneven matrix incorporation and lethality
Collapse
Affiliation(s)
- Yutaka Matsubayashi
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK.
| | - Adam Louani
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Anca Dragu
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | | | - Eduardo Serna-Morales
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Lawrence Yolland
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Attila Gyoergy
- Institute of Science and Technology, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Gema Vizcay
- Centre for Ultrastructure Imaging, King's College London, London SE1 1UL, UK
| | - Roland A Fleck
- Centre for Ultrastructure Imaging, King's College London, London SE1 1UL, UK
| | - John M Heddleston
- Advanced Imaging Center, Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | - Teng-Leong Chew
- Advanced Imaging Center, Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | - Daria E Siekhaus
- Institute of Science and Technology, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Brian M Stramer
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK.
| |
Collapse
|
48
|
Chioran A, Duncan S, Catalano A, Brown TJ, Ringuette MJ. Collagen IV trafficking: The inside-out and beyond story. Dev Biol 2017; 431:124-133. [PMID: 28982537 DOI: 10.1016/j.ydbio.2017.09.037] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 09/28/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022]
Abstract
Collagen IV networks endow basement membranes (BMs) with remarkable tensile strength and function as morphoregulatory substrata for diverse tissue-specific developmental events. A complex repertoire of intracellular and extracellular molecular interactions are required for collagen IV secretion and supramolecular assembly into BMs. These include intracellular chaperones such as Heat shock protein 47 (Hsp47) and the chaperone-binding trafficking protein Transport and Golgi organization protein 1 (Tango1). Mutations in these proteins lead to compromised collagen IV protomer stability and secretion, leading to defective BM assembly and function. In addition to intracellular chaperones, a role for extracellular chaperones orchestrating the transport, supramolecular assembly, and architecture of collagen IV in BM is emerging. We present evidence derived from evolutionarily distant model organisms that supports an extracellular collagen IV chaperone-like activity for the matricellular protein SPARC (Secreted Protein, Acidic, Rich in Cysteine). Loss of SPARC disrupts BM homeostasis and compromises tissue biomechanics and physiological function. Thus, the combined contributions of intracellular and extracellular collagen IV-associated chaperones and chaperone-like proteins are critical to ensure proper secretion and stereotypic assembly of collagen IV networks in BMs.
Collapse
Affiliation(s)
- Alexa Chioran
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada M5S 3G5
| | - Sebastian Duncan
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada M5S 3G5
| | | | - Theodore J Brown
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, ON, Canada
| | - Maurice J Ringuette
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada M5S 3G5.
| |
Collapse
|
49
|
Live-cell confocal microscopy and quantitative 4D image analysis of anchor-cell invasion through the basement membrane in Caenorhabditis elegans. Nat Protoc 2017; 12:2081-2096. [PMID: 28880279 DOI: 10.1038/nprot.2017.093] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cell invasion through basement membrane (BM) barriers is crucial in development, leukocyte trafficking and the spread of cancer. The mechanisms that direct invasion, despite their importance in normal and disease states, are poorly understood, largely because of the inability to visualize dynamic cell-BM interactions in vivo. This protocol describes multichannel time-lapse confocal imaging of anchor-cell invasion in live Caenorhabditis elegans. Methods presented include outline-slide preparation and worm growth synchronization (15 min), mounting (20 min), image acquisition (20-180 min), image processing (20 min) and quantitative analysis (variable timing). The acquired images enable direct measurement of invasive dynamics including formation of invadopodia and cell-membrane protrusions, and removal of BM. This protocol can be combined with genetic analysis, molecular-activity probes and optogenetic approaches to uncover the molecular mechanisms underlying cell invasion. These methods can also be readily adapted by any worm laboratory for real-time analysis of cell migration, BM turnover and cell-membrane dynamics.
Collapse
|
50
|
Blockhuys S, Wittung-Stafshede P. Roles of Copper-Binding Proteins in Breast Cancer. Int J Mol Sci 2017; 18:ijms18040871. [PMID: 28425924 PMCID: PMC5412452 DOI: 10.3390/ijms18040871] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/11/2017] [Accepted: 04/18/2017] [Indexed: 12/17/2022] Open
Abstract
Copper ions are needed in several steps of cancer progression. However, the underlying mechanisms, and involved copper-binding proteins, are mainly elusive. Since most copper ions in the body (in and outside cells) are protein-bound, it is important to investigate what copper-binding proteins participate and, for these, how they are loaded with copper by copper transport proteins. Mechanistic information for how some copper-binding proteins, such as extracellular lysyl oxidase (LOX), play roles in cancer have been elucidated but there is still much to learn from a biophysical molecular viewpoint. Here we provide a summary of copper-binding proteins and discuss ones reported to have roles in cancer. We specifically focus on how copper-binding proteins such as mediator of cell motility 1 (MEMO1), LOX, LOX-like proteins, and secreted protein acidic and rich in cysteine (SPARC) modulate breast cancer from molecular and clinical aspects. Because of the importance of copper for invasion/migration processes, which are key components of cancer metastasis, further insights into the actions of copper-binding proteins may provide new targets to combat cancer.
Collapse
Affiliation(s)
- Stéphanie Blockhuys
- Department Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden.
| | - Pernilla Wittung-Stafshede
- Department Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden.
| |
Collapse
|