1
|
Botelho CM, Rodríguez-Cabello JC, Barbosa MA. Harnessing Thrombospondin-1-Enabled Decellularized Nucleus Pulposus Matrices and Elastin-Like Recombinamers to Rebuild an Avascular Analogue of the Intervertebral Disc. J Biomed Mater Res A 2025; 113:e37911. [PMID: 40269577 DOI: 10.1002/jbm.a.37911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025]
Abstract
With the degeneration of the intervertebral disc (IVD), the ingrowth of vascular and neural structures occurs. Both nerves and blood vessels engage in the development of inflammation and the onset of discogenic pain. The present study aimed to produce a hierarchical biomaterial capable of inhibiting angiogenesis by emulating the microenvironment of non-degenerated IVDs. To this end, we have incorporated an angiogenesis modulator-thrombospondin-1 (TSP-1) into a three-dimensional (3D) hydrogel network containing decellularized nucleus pulposus (dNPs) and azide-cyclooctyne modified elastin-like recombinamers (ELRs). Following the decellularization of nucleus pulposus (NPs) isolated from bovine tissues, pre-gels (pGs) were assembled based on the acid-pepsin extraction of soluble collagens found in the dNPs. Given the inherent affinity of these macromolecules to TSP-1, which was corroborated by immunohistochemical analysis and FT-IR spectroscopy, the pGs were supplemented with two concentrations of TSP-1. Angiogenesis was evaluated using the chick chorioallantoic membrane (CAM) in vivo model. Conjugation of TSP-1 with the pGs resulted in a synergistic suppression of blood vessel formation. Complexation with the ELRs improved the viscoelastic moduli and the structural stability of the hydrogels, which maintained their hydration and osmolarity properties due to the presence of the dNPs. When placed in direct contact with human primary fibroblasts, the materials displayed high cytocompatibility and tunable degradation rates. Our findings indicate that TSP-1-enabled dNP-derived pGs inhibit angiogenesis in vivo, while the presence of the ELRs aids in improving the mechanical properties of the hydrogels, thus providing a platform for rebuilding an avascular analogue of the healthy IVD.
Collapse
Affiliation(s)
- Carlos Marinho Botelho
- i3S - Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - José Carlos Rodríguez-Cabello
- BIOFORGE - Grupo de Materiales Avanzados y Nanobiotecnología, Universidad de Valladolid, L.A.D.I.S., CIBER-BBN, Valladolid, Spain
| | - Mário Adolfo Barbosa
- i3S - Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
2
|
Dudek P, Talar-Wojnarowska R. Current Approach to Risk Factors and Biomarkers of Intestinal Fibrosis in Inflammatory Bowel Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:305. [PMID: 38399592 PMCID: PMC10889938 DOI: 10.3390/medicina60020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Inflammatory bowel disease (IBD), especially Crohn's disease (CD), characterized by a chronic inflammatory process and progressive intestinal tissue damage, leads to the unrestrained proliferation of mesenchymal cells and the development of bowel strictures. Complications induced by fibrosis are related to high rates of morbidity and mortality and lead to a substantial number of hospitalizations and surgical procedures, generating high healthcare costs. The development of easily obtained, reliable fibrogenesis biomarkers is essential to provide an important complementary tool to existing diagnostic and prognostic methods in IBD management, guiding decisions on the intensification of pharmacotherapy, proceeding to surgical methods of treatment and monitoring the efficacy of anti-fibrotic therapy in the future. The most promising potential markers of fibrosis include cartilage oligomeric matrix protein (COMP), hepatocyte growth factor activator (HGFA), and fibronectin isoform- extra domain A (ED-A), as well as antibodies against granulocyte macrophage colony-stimulating factor (GM-CSF Ab), cathelicidin (LL-37), or circulatory miRNAs: miR-19a-3p and miR-19b-3p. This review summarizes the role of genetic predisposition, and risk factors and serological markers potentially contributing to the pathophysiology of fibrotic strictures in the course of IBD.
Collapse
|
3
|
South AP, Laimer M, Gueye M, Sui JY, Eichenfield LF, Mellerio JE, Nyström A. Type VII Collagen Deficiency in the Oncogenesis of Cutaneous Squamous Cell Carcinoma in Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2023; 143:2108-2119. [PMID: 37327859 DOI: 10.1016/j.jid.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Dystrophic epidermolysis bullosa is a rare genetic skin disorder caused by COL7A1 sequence variations that result in type VII collagen deficits and cutaneous and extracutaneous manifestations. One serious complication of dystrophic epidermolysis bullosa is cutaneous squamous cell carcinoma, a leading driver of morbidity and mortality, especially among patients with recessive dystrophic epidermolysis bullosa. Type VII collagen deficits alter TGFβ signaling and evoke multiple other cutaneous squamous cell carcinoma progression-promoting activities within epidermal microenvironments. This review examines cutaneous squamous cell carcinoma pathophysiology in dystrophic epidermolysis bullosa with a focus on known oncogenesis pathways at play and explores the idea that therapeutic type VII collagen replacement may reduce cutaneous squamous cell carcinoma risk.
Collapse
Affiliation(s)
- Andrew P South
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Martin Laimer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | | | - Jennifer Y Sui
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Jemima E Mellerio
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, Freiburg, Germany; Freiburg Institute for Advanced Studies, Freiburg, Germany
| |
Collapse
|
4
|
Sano T, Ochiai T, Nagayama T, Nakamura A, Kubota N, Kadowaki T, Wakabayashi T, Iwatsubo T. Genetic Reduction of Insulin Signaling Mitigates Amyloid-β Deposition by Promoting Expression of Extracellular Matrix Proteins in the Brain. J Neurosci 2023; 43:7226-7241. [PMID: 37699718 PMCID: PMC10601373 DOI: 10.1523/jneurosci.0071-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/16/2023] [Accepted: 09/03/2023] [Indexed: 09/14/2023] Open
Abstract
The insulin/IGF-1 signaling (IIS) regulates a wide range of biological processes, including aging and lifespan, and has also been implicated in the pathogenesis of Alzheimer's disease (AD). We and others have reported that reduced signaling by genetic ablation of the molecules involved in IIS (e.g., insulin receptor substrate 2 [IRS-2]) markedly mitigates amyloid plaque formation in the brains of mouse models of AD, although the molecular underpinnings of the amelioration remain unsolved. Here, we revealed, by a transcriptomic analysis of the male murine cerebral cortices, that the expression of genes encoding extracellular matrix (ECM) was significantly upregulated by the loss of IRS-2. Insulin signaling activity negatively regulated the phosphorylation of Smad2 and Smad3 in the brain, and suppressed TGF-β/Smad-dependent expression of a subset of ECM genes in brain-derived cells. The ECM proteins inhibited Aβ fibril formation in vitro, and IRS-2 deficiency suppressed the aggregation process of Aβ in the brains of male APP transgenic mice as revealed by injection of aggregation seeds in vivo Our results propose a novel mechanism in AD pathophysiology whereby IIS modifies Aβ aggregation and amyloid pathology by altering the expression of ECM genes in the brain.SIGNIFICANCE STATEMENT The insulin/IGF-1 signaling (IIS) has been recognized as a regulator of aging, a leading risk factor for the onset of Alzheimer's disease (AD). In AD mouse models, genetic deletion of key IIS molecules markedly reduces the amyloid plaque formation in the brain, although the molecular underpinnings of this amelioration remain elusive. We found that the deficiency of insulin receptor substrate 2 leads to an increase in the expression of various extracellular matrices (ECMs) in the brain, potentially through TGF-β/Smad signaling. Furthermore, some of those ECMs exhibited the potential to inhibit amyloid plaque accumulation by disrupting the formation of Aβ fibrils. This study presents a novel mechanism by which IIS regulates Aβ accumulation, which may involve altered brain ECM expression.
Collapse
Affiliation(s)
- Toshiharu Sano
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Toshitaka Ochiai
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- Pharmacology Department, Drug Research Center, Kaken Pharmaceutical Company, LTD, Kyoto, 607-8042, Japan
| | - Takeru Nagayama
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Ayaka Nakamura
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- Department of Clinical Nutrition Therapy, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- Toranomon Hospital, Tokyo, 105-8470, Japan
| | - Tomoko Wakabayashi
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- Department of Innovative Dementia Prevention, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
5
|
Dolmatov IY, Nizhnichenko VA. Extracellular Matrix of Echinoderms. Mar Drugs 2023; 21:417. [PMID: 37504948 PMCID: PMC10381214 DOI: 10.3390/md21070417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
This review considers available data on the composition of the extracellular matrix (ECM) in echinoderms. The connective tissue in these animals has a rather complex organization. It includes a wide range of structural ECM proteins, as well as various proteases and their inhibitors. Members of almost all major groups of collagens, various glycoproteins, and proteoglycans have been found in echinoderms. There are enzymes for the synthesis of structural proteins and their modification by polysaccharides. However, the ECM of echinoderms substantially differs from that of vertebrates by the lack of elastin, fibronectins, tenascins, and some other glycoproteins and proteoglycans. Echinoderms have a wide variety of proteinases, with serine, cysteine, aspartic, and metal peptidases identified among them. Their active centers have a typical structure and can break down various ECM molecules. Echinoderms are also distinguished by a wide range of proteinase inhibitors. The complex ECM structure and the variety of intermolecular interactions evidently explain the complexity of the mechanisms responsible for variations in the mechanical properties of connective tissue in echinoderms. These mechanisms probably depend not only on the number of cross-links between the molecules, but also on the composition of ECM and the properties of its proteins.
Collapse
Affiliation(s)
- Igor Yu Dolmatov
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevsky 17, 690041 Vladivostok, Russia
| | - Vladimir A Nizhnichenko
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevsky 17, 690041 Vladivostok, Russia
| |
Collapse
|
6
|
Fabian O, Bajer L, Drastich P, Harant K, Sticova E, Daskova N, Modos I, Tichanek F, Cahova M. A Current State of Proteomics in Adult and Pediatric Inflammatory Bowel Diseases: A Systematic Search and Review. Int J Mol Sci 2023; 24:ijms24119386. [PMID: 37298338 DOI: 10.3390/ijms24119386] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are systemic immune-mediated conditions with predilection for the gastrointestinal tract and include Crohn's disease and ulcerative colitis. Despite the advances in the fields of basic and applied research, the etiopathogenesis remains largely unknown. As a result, only one third of the patients achieve endoscopic remission. A substantial portion of the patients also develop severe clinical complications or neoplasia. The need for novel biomarkers that can enhance diagnostic accuracy, more precisely reflect disease activity, and predict a complicated disease course, thus, remains high. Genomic and transcriptomic studies contributed substantially to our understanding of the immunopathological pathways involved in disease initiation and progression. However, eventual genomic alterations do not necessarily translate into the final clinical picture. Proteomics may represent a missing link between the genome, transcriptome, and phenotypical presentation of the disease. Based on the analysis of a large spectrum of proteins in tissues, it seems to be a promising method for the identification of new biomarkers. This systematic search and review summarize the current state of proteomics in human IBD. It comments on the utility of proteomics in research, describes the basic proteomic techniques, and provides an up-to-date overview of available studies in both adult and pediatric IBD.
Collapse
Affiliation(s)
- Ondrej Fabian
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
- Department of Pathology and Molecular Medicine, 3rd Faculty of Medicine, Charles University and Thomayer Hospital, 140 59 Prague, Czech Republic
| | - Lukas Bajer
- Department of Gastroenterology and Hepatology, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
- Institute of Microbiology, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Pavel Drastich
- Department of Gastroenterology and Hepatology, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | - Karel Harant
- Proteomics Core Facility, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Eva Sticova
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
- Department of Pathology, Royal Vinohrady Teaching Hospital, Srobarova 1150/50, 100 00 Prague, Czech Republic
| | - Nikola Daskova
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | - Istvan Modos
- Department of Informatics, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | - Filip Tichanek
- Department of Informatics, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | - Monika Cahova
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| |
Collapse
|
7
|
Gao G, Li X, Jiang Z, Osorio L, Tang YL, Yu X, Jin G, Zhou Z. Isthmin-1 (Ism1) modulates renal branching morphogenesis and mesenchyme condensation during early kidney development. Nat Commun 2023; 14:2378. [PMID: 37185772 PMCID: PMC10130008 DOI: 10.1038/s41467-023-37992-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
The outgrowth of epithelial bud followed by reiterated bifurcations during renal development is driven by the ligand-receptor interactions between the epithelium and the surrounding mesenchyme. Here, by exploring ligand-receptor interactions in E10.5 and E11.5 kidneys by single cell RNA-seq, we find that Isthmin1 (Ism1), a secreted protein, resembles Gdnf expression and modulates kidney branching morphogenesis. Mice deficient for Ism1 exhibit defective ureteric bud bifurcation and impaired metanephric mesenchyme condensation in E11.5 embryos, attributable to the compromised Gdnf/Ret signaling, ultimately leading to renal agenesis and hypoplasia/dysplasia. By HRP-induced proximity labelling, we further identify integrin α8β1 as a receptor of Ism1 in E11.5 kidney and demonstrate that Ism1 promoted cell-cell adhesion through interacting with Integrin α8β1, the receptor whose activation is responsible for Gdnf expression and mesenchyme condensation. Taken together, our work reveals Ism1 as a critical regulator of cell-cell interaction that modulates Gdnf/Ret signaling during early kidney development.
Collapse
Affiliation(s)
- Ge Gao
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
| | - Zhixin Jiang
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China
| | - Liliana Osorio
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China
| | - Ying Lam Tang
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China
| | - Xueqing Yu
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Guoxiang Jin
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Zhongjun Zhou
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China.
- Reproductive Medical Center, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
8
|
Peserico A, Barboni B, Russo V, Bernabò N, El Khatib M, Prencipe G, Cerveró-Varona A, Haidar-Montes AA, Faydaver M, Citeroni MR, Berardinelli P, Mauro A. Mammal comparative tendon biology: advances in regulatory mechanisms through a computational modeling. Front Vet Sci 2023; 10:1175346. [PMID: 37180059 PMCID: PMC10174257 DOI: 10.3389/fvets.2023.1175346] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023] Open
Abstract
There is high clinical demand for the resolution of tendinopathies, which affect mainly adult individuals and animals. Tendon damage resolution during the adult lifetime is not as effective as in earlier stages where complete restoration of tendon structure and property occurs. However, the molecular mechanisms underlying tendon regeneration remain unknown, limiting the development of targeted therapies. The research aim was to draw a comparative map of molecules that control tenogenesis and to exploit systems biology to model their signaling cascades and physiological paths. Using current literature data on molecular interactions in early tendon development, species-specific data collections were created. Then, computational analysis was used to construct Tendon NETworks in which information flow and molecular links were traced, prioritized, and enriched. Species-specific Tendon NETworks generated a data-driven computational framework based on three operative levels and a stage-dependent set of molecules and interactions (embryo-fetal or prepubertal) responsible, respectively, for signaling differentiation and morphogenesis, shaping tendon transcriptional program and downstream modeling of its fibrillogenesis toward a mature tissue. The computational network enrichment unveiled a more complex hierarchical organization of molecule interactions assigning a central role to neuro and endocrine axes which are novel and only partially explored systems for tenogenesis. Overall, this study emphasizes the value of system biology in linking the currently available disjointed molecular data, by establishing the direction and priority of signaling flows. Simultaneously, computational enrichment was critical in revealing new nodes and pathways to watch out for in promoting biomedical advances in tendon healing and developing targeted therapeutic strategies to improve current clinical interventions.
Collapse
|
9
|
Thamrongwaranggoon U, Kuribayashi K, Araki H, Hino Y, Koga T, Seubwai W, Wongkham S, Nakao M, Hino S. Lactic acidosis induces metabolic and phenotypic reprogramming in cholangiocarcinoma cells via the upregulation of thrombospondin-1. Cancer Sci 2022; 114:1541-1555. [PMID: 36562400 PMCID: PMC10067391 DOI: 10.1111/cas.15699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
The high glycolytic activity of cancer cells leads to lactic acidosis (LA) in the tumor microenvironment. LA is not merely a consequence of metabolic activities but also has functional roles in metabolic reprogramming and cancer progression. Cholangiocarcinoma (CCA) cells exhibit a high dependency on glycolysis for survival and growth, but the specific effects of LA on cellular characteristics remain unknown. Here, we demonstrate that long-term LA (LLA) reprograms the metabolic phenotype of CCA cells from glycolytic to oxidative and enhances their migratory activity. In CCA cell culture, short-term LA (24 h) showed a growth inhibitory effect, while extended LA exposure for more than 2 weeks (LLA) led to enhanced cell motility. Coincidentally, LLA enhanced the respiratory capacity with an increase in mitochondrial mass. Inhibition of mitochondrial function abolished LLA-induced cell motility, suggesting that metabolic remodeling affects the phenotypic outcomes. RNA-sequencing analysis revealed that LLA upregulated genes associated with cell migration and epithelial-mesenchymal transition (EMT), including thrombospondin-1 (THBS1), which encodes a pro-EMT-secreted protein. Inhibition of THBS1 resulted in the suppression of both LLA-induced cell motility and respiratory capacity. Moreover, high THBS1 expression was associated with poor survival in patients with CCA. Collectively, our study suggests that the increased expression of THBS1 by LLA promotes phenotypic alterations, leading to CCA progression.
Collapse
Affiliation(s)
- Ubonrat Thamrongwaranggoon
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kanji Kuribayashi
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Hirotaka Araki
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Yuko Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Tomoaki Koga
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Wunchana Seubwai
- Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Department of Forensic Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
10
|
Biopolymer-Based Wound Dressings with Biochemical Cues for Cell-Instructive Wound Repair. Polymers (Basel) 2022; 14:polym14245371. [PMID: 36559739 PMCID: PMC9783382 DOI: 10.3390/polym14245371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/25/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine is an active research sphere that focuses on the repair, regeneration, and replacement of damaged tissues and organs. A plethora of innovative wound dressings and skin substitutes have been developed to treat cutaneous wounds and are aimed at reducing the length or need for a hospital stay. The inception of biomaterials with the ability to interact with cells and direct them toward desired lineages has brought about innovative designs in wound healing and tissue engineering. This cellular engagement is achieved by cell cues that can be biochemical or biophysical in nature. In effect, these cues seep into innate repair pathways, cause downstream cell behaviours and, ultimately, lead to advantageous healing. This review will focus on biomolecules with encoded biomimetic, instructive prompts that elicit desired cellular domino effects to achieve advanced wound repair. The wound healing dressings covered in this review are based on functionalized biopolymeric materials. While both biophysical and biochemical cues are vital for advanced wound healing applications, focus will be placed on biochemical cues and in vivo or clinical trial applications. The biochemical cues aforementioned will include peptide therapy, collagen matrices, cell-based therapy, decellularized matrices, platelet-rich plasma, and biometals.
Collapse
|
11
|
Cárdenas-León CG, Mäemets-Allas K, Klaas M, Lagus H, Kankuri E, Jaks V. Matricellular proteins in cutaneous wound healing. Front Cell Dev Biol 2022; 10:1073320. [PMID: 36506087 PMCID: PMC9730256 DOI: 10.3389/fcell.2022.1073320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Cutaneous wound healing is a complex process that encompasses alterations in all aspects of the skin including the extracellular matrix (ECM). ECM consist of large structural proteins such as collagens and elastin as well as smaller proteins with mainly regulative properties called matricellular proteins. Matricellular proteins bind to structural proteins and their functions include but are not limited to interaction with cell surface receptors, cytokines, or protease and evoking a cellular response. The signaling initiated by matricellular proteins modulates differentiation and proliferation of cells having an impact on the tissue regeneration. In this review we give an overview of the matricellular proteins that have been found to be involved in cutaneous wound healing and summarize the information known to date about their functions in this process.
Collapse
Affiliation(s)
| | - Kristina Mäemets-Allas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Mariliis Klaas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Heli Lagus
- Department of Plastic Surgery and Wound Healing Centre, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Viljar Jaks
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia,Dermatology Clinic, Tartu University Clinics, Tartu, Estonia,*Correspondence: Viljar Jaks,
| |
Collapse
|
12
|
Tabary M, Gheware A, Peñaloza HF, Lee JS. The matricellular protein thrombospondin-1 in lung inflammation and injury. Am J Physiol Cell Physiol 2022; 323:C857-C865. [PMID: 35912991 PMCID: PMC9467471 DOI: 10.1152/ajpcell.00182.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/22/2022]
Abstract
Matricellular proteins comprise a diverse group of molecular entities secreted into the extracellular space. They interact with the extracellular matrix (ECM), integrins, and other cell-surface receptors, and can alter matrix strength, cell attachment to the matrix, and cell-cell adhesion. A founding member of this group is thrombospondin-1 (TSP-1), a high molecular-mass homotrimeric glycoprotein. Given the importance of the matrix and ECM remodeling in the lung following injury, TSP-1 has been implicated in a number of lung pathologies. This review examines the role of TSP-1 as a damage controller in the context of lung inflammation, injury resolution, and repair in noninfectious and infectious models. This review also discusses the potential role of TSP-1 in human diseases as it relates to lung inflammation and injury.
Collapse
Affiliation(s)
- Mohammadreza Tabary
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Atish Gheware
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hernán F Peñaloza
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Janet S Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
13
|
Cartilage Oligomeric Matrix Protein, Diseases, and Therapeutic Opportunities. Int J Mol Sci 2022; 23:ijms23169253. [PMID: 36012514 PMCID: PMC9408827 DOI: 10.3390/ijms23169253] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cartilage oligomeric matrix protein (COMP) is an extracellular matrix (ECM) glycoprotein that is critical for collagen assembly and ECM stability. Mutations of COMP cause endoplasmic reticulum stress and chondrocyte apoptosis, resulting in rare skeleton diseases. The bouquet-like structure of COMP allows it to act as a bridging molecule that regulates cellular phenotype and function. COMP is able to interact with many other ECM components and binds directly to a variety of cellular receptors and growth factors. The roles of COMP in other skeleton diseases, such as osteoarthritis, have been implied. As a well-established biochemical marker, COMP indicates cartilage turnover associated with destruction. Recent exciting achievements indicate its involvement in other diseases, such as malignancy, cardiovascular diseases, and tissue fibrosis. Here, we review the basic concepts of COMP and summarize its novel functions in the regulation of signaling events. These findings renew our understanding that COMP has a notable function in cell behavior and disease progression as a signaling regulator. Interestingly, COMP shows distinct functions in different diseases. Targeting COMP in malignancy may withdraw its beneficial effects on the vascular system and induce or aggravate cardiovascular diseases. COMP supplementation is a promising treatment for OA and aortic aneurysms while it may induce tissue fibrosis or cancer metastasis.
Collapse
|
14
|
Taglietti V, Kefi K, Bronisz-Budzyńska I, Mirciloglu B, Rodrigues M, Cardone N, Coulpier F, Periou B, Gentil C, Goddard M, Authier FJ, Pietri-Rouxel F, Malfatti E, Lafuste P, Tiret L, Relaix F. Duchenne muscular dystrophy trajectory in R-DMDdel52 preclinical rat model identifies COMP as biomarker of fibrosis. Acta Neuropathol Commun 2022; 10:60. [PMID: 35468843 PMCID: PMC9036715 DOI: 10.1186/s40478-022-01355-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/25/2022] [Indexed: 11/10/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disorder caused by mutations in the Dystrophin gene and for which there is currently no cure. To bridge the gap between preclinical and therapeutic evaluation studies, we have generated a rat model for DMD that carries an exon 52 deletion (R-DMDdel52) causing a complete lack of dystrophin protein. Here we show that R-DMDdel52 animals recapitulated human DMD pathophysiological trajectory more faithfully than the mdx mouse model. We report that R-DMDdel52 rats displayed progressive and severe skeletal muscle loss associated with fibrotic deposition, fat infiltration and fibre type switch. Early fibrosis was also apparent in the cardiac muscle. These histological modifications led to severe muscle, respiratory and cardiac functional impairments leading to premature death around 1 year. Moreover, DMD muscle exhibited systemic inflammation with a mixed M1/M2 phenotype. A comparative single cell RNAseq analysis of the diaphragm muscle was performed, revealing cellular populations alteration and molecular modifications in all muscle cell types. We show that DMD fibroadipogenic progenitors produced elevated levels of cartilage oligomeric matrix protein, a glycoprotein responsible for modulating homeostasis of extracellular matrix, and whose increased concentration correlated with muscle fibrosis both in R-DMDdel52 rats and human patients. Fibrosis is a component of tissue remodelling impacting the whole musculature of DMD patients, at the tissue level but most importantly at the functional level. We therefore propose that this specific biomarker can optimize the prognostic monitoring of functional improvement of patients included in clinical trials.
Collapse
|
15
|
Single-cell RNA sequencing identifies a migratory keratinocyte subpopulation expressing THBS1 in epidermal wound healing. iScience 2022; 25:104130. [PMID: 35391830 PMCID: PMC8980802 DOI: 10.1016/j.isci.2022.104130] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/19/2021] [Accepted: 03/17/2022] [Indexed: 11/30/2022] Open
Abstract
Keratinocyte differentiation is an intricate process that is regulated by multiple mediators. Using cultured human keratinocytes, we found that lysophosphatidic acid (LPA) induced the differentiation of a previously unsuspected keratinocyte subpopulation expressing the extracellular matrix protein, thrombospondin-1 (THBS1). This action of LPA was mediated by the RHO/ROCK-SRF signaling downstream of LPA1 and LPA5 receptors and required ERK activity. Suppression of THBS1 in vitro suggested a migratory role of THBS1+ keratinocytes. Moreover, we analyzed publicly deposited single-cell RNA sequencing dataset and identified Thbs1-expressing keratinocytes in the mouse wound skin. Immunohistochemistry analysis revealed that Thbs1+ keratinocytes were apparently differentiated from basal keratinocytes upon wounding, subsequently polarized and migrated suprabasally toward the wound front, and eventually underwent terminal differentiation in the neo-epidermis. Importantly, inhibition of Erk activity suppressed Thbs1+ keratinocyte differentiation in wound healing. Based on these findings, we suggest that THBS1+ keratinocyte is a migratory keratinocyte subpopulation that facilitates epidermal wound healing. Single-cell RNA sequencing reveals a keratinocyte subpopulation expressing THBS1 LPA and ERK activity are required for the induction of THBS1+ keratinocyte Thbs1+ keratinocytes are differentiated from basal keratinocytes upon epidermal wounding Thbs1+ keratinocytes are migratory cells and may contribute to epidermal wound healing
Collapse
|
16
|
Plana E, Oto J, Medina P, Herranz R, Fernández-Pardo Á, Requejo L, Miralles M. Thrombospondins in human aortic aneurysms. IUBMB Life 2022; 74:982-994. [PMID: 35293116 DOI: 10.1002/iub.2610] [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: 09/03/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 11/08/2022]
Abstract
Thrombospondins are a family of matricellular proteins with a multimeric structure that is known to be involved in several biological and pathological processes. Their relationship with vascular disorders has raised special interest recently. Aortic aneurysms are related to the impairment of vascular remodeling, in which extracellular matrix proteins seem to play an important role. Thus, research in thrombospondins, and their potential role in aneurysm development is progressively gaining importance. Nevertheless, studies showing thrombospondin dysregulation in human samples are still scarce. Although studies performed in vitro and in vivo models are essential to understand the molecular mechanisms and pathways underlying the disorder, descriptive studies in human samples are also necessary to ascertain their real value as biomarkers and/or novel therapeutic targets. The present article reviews the latest findings regarding the role of thrombospondins in aortic aneurysm development, paying particular attention to the studies performed in human samples.
Collapse
Affiliation(s)
- Emma Plana
- Angiology and Vascular Surgery Service, La Fe University and Polytechnic Hospital, Valencia, Spain.,Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Julia Oto
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Pilar Medina
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Raquel Herranz
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Álvaro Fernández-Pardo
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Lucia Requejo
- Angiology and Vascular Surgery Service, La Ribera University Hospital, Alzira, Valencia, Spain
| | - Manuel Miralles
- Angiology and Vascular Surgery Service, La Fe University and Polytechnic Hospital, Valencia, Spain.,Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain.,Department of Surgery, University of Valencia, Valencia, Spain
| |
Collapse
|
17
|
Arun A, Rayford KJ, Cooley A, Rana T, Rachakonda G, Villalta F, Pratap S, Lima MF, Sheibani N, Nde PN. Thrombospondin-1 expression and modulation of Wnt and hippo signaling pathways during the early phase of Trypanosoma cruzi infection of heart endothelial cells. PLoS Negl Trop Dis 2022; 16:e0010074. [PMID: 34986160 PMCID: PMC8730400 DOI: 10.1371/journal.pntd.0010074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
The protozoan parasite, Trypanosoma cruzi, causes severe morbidity and mortality in afflicted individuals. Approximately 30% of T. cruzi infected individuals present with cardiac pathology. The invasive forms of the parasite are carried in the vascular system to infect other cells of the body. During transportation, the molecular mechanisms by which the parasite signals and interact with host endothelial cells (EC) especially heart endothelium is currently unknown. The parasite increases host thrombospondin-1 (TSP1) expression and activates the Wnt/β-catenin and hippo signaling pathways during the early phase of infection. The links between TSP1 and activation of the signaling pathways and their impact on parasite infectivity during the early phase of infection remain unknown. To elucidate the significance of TSP1 function in YAP/β-catenin colocalization and how they impact parasite infectivity during the early phase of infection, we challenged mouse heart endothelial cells (MHEC) from wild type (WT) and TSP1 knockout mice with T. cruzi and evaluated Wnt signaling, YAP/β-catenin crosstalk, and how they affect parasite infection. We found that in the absence of TSP1, the parasite induced the expression of Wnt-5a to a maximum at 2 h (1.73±0.13), P< 0.001 and enhanced the level of phosphorylated glycogen synthase kinase 3β at the same time point (2.99±0.24), P<0.001. In WT MHEC, the levels of Wnt-5a were toned down and the level of p-GSK-3β was lowest at 2 h (0.47±0.06), P< 0.01 compared to uninfected control. This was accompanied by a continuous significant increase in the nuclear colocalization of β-catenin/YAP in TSP1 KO MHEC with a maximum Pearson correlation coefficient of (0.67±0.02), P< 0.05 at 6 h. In WT MHEC, the nuclear colocalization of β-catenin/YAP remained steady and showed a reduction at 6 h (0.29±0.007), P< 0.05. These results indicate that TSP1 plays an important role in regulating β-catenin/YAP colocalization during the early phase of T. cruzi infection. Importantly, dysregulation of this crosstalk by pre-incubation of WT MHEC with a β-catenin inhibitor, endo-IWR 1, dramatically reduced the level of infection of WT MHEC. Parasite infectivity of inhibitor treated WT MHEC was similar to the level of infection of TSP1 KO MHEC. These results indicate that the β-catenin pathway induced by the parasite and regulated by TSP1 during the early phase of T. cruzi infection is an important potential therapeutic target, which can be explored for the prophylactic prevention of T. cruzi infection.
Collapse
Affiliation(s)
- Ashutosh Arun
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Ayorinde Cooley
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Tanu Rana
- Department of Professional Medical Education and Molecular Biology Core Facility, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Girish Rachakonda
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Fernando Villalta
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Siddharth Pratap
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Maria F. Lima
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, United States of America
- Department of Molecular and Cellular and Biomedical Sciences, School of Medicine, The City College of New York, New York, United States of America
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| |
Collapse
|
18
|
Chae DS, Kim ME, Kang KY, Lee NY, Lee WS, Lee JS. Quantitative proteomic analysis comparing grades ICRS1 and ICRS3 in patients with osteoarthritis. Exp Ther Med 2021; 22:1470. [PMID: 34737810 PMCID: PMC8561757 DOI: 10.3892/etm.2021.10905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/07/2021] [Indexed: 11/06/2022] Open
Abstract
Osteoarthritis (OA), which is caused by joint damage, is the most common form of arthritis, affecting millions of people worldwide. This damage can accumulate over time, which is why aging is one of the main contributors to joint damage associated with OA. The OA-related proteins that have been reported to date have been identified by the comparative analysis of OA patients with normal controls, following surgical or pharmacological treatment. For the first time, the present study analyzed OA-related proteins in patients with OA according to the International Cartilage Repair Society (ICRS) scale. Changes in protein expression can be observed during the OA process. The present study demonstrated differential protein expression patterns in articular cartilage from ICRS1- and ICRS3-graded OA patients. ICRS grade-matched OA knee samples from 12 OA patients, 6 ICRS grade 1 patients and 6 ICRS3 patients were subjected to proteomic analysis using the LTQ-Orbitrap mass spectrometry system. A total of 231 unique proteins were identified as expressed across the ICRS1 and ICRS3 OA patient groups. Relative differences in protein expression associated with the following classifications were observed: Biological adhesion, cell killing, cellular process, development process and molecular function. Although some of these proteins have been previously reported to be associated with rheumatoid arthritis, including cartilage oligomeric matrix protein, collagen types, angiogenin, complement C5 and CD59 glycoprotein, numerous additional proteins were newly identified, which may further help our understanding of disease pathogenesis. These findings suggested that these proteins may be used to develop novel therapeutic targets for OA.
Collapse
Affiliation(s)
- Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary's Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Mi Eun Kim
- Department of Life Science, Immunology Research Lab, BK21-Four Education Research Group, College of Natural Sciences, Chosun University, Gwangju 61452, Republic of Korea
| | - Kyung-Yil Kang
- Department of Orthopedic Surgery, International St. Mary's Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Nae Yoon Lee
- Department of Bionano Technology, Gachon University, Seongnam, Gyeonggi-do 13120, Republic of Korea
| | - Woo-Suk Lee
- Department of Orthopedic Surgery, Yonsei University College of Medicine, Gangnam Severence Hospital, Seoul 135720, Republic of Korea
| | - Jun Sik Lee
- Department of Life Science, Immunology Research Lab, BK21-Four Education Research Group, College of Natural Sciences, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
19
|
Zhang S, Sharaf Eldin HE, Gu WL, Li TS. Laminin alpha-3 and thrombospondin-1 differently regulate the survival and differentiation of hepatocytes and hepatic stem cells from neonatal mice. Am J Transl Res 2021; 13:12684-12693. [PMID: 34956483 PMCID: PMC8661240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 03/31/2021] [Indexed: 06/14/2023]
Abstract
The aim of this study was to search and identify the extracellular matrix/adhesion molecules potentially regulating liver regeneration. By using pathway-focused PCR array, we investigated the dynamic changes in the expression of extracellular matrix and adhesion molecules in normal livers or cholestatic livers following partial hepatectomy in adult mice. To confirm the data from PCR array, we further evaluated how laminin alpha-3 and thrombospondin-1 mediate the survival and differentiation of matured hepatocytes and immature hepatic stem cells by using primarily isolated liver cells from neonatal mice. According to the different changes in the expression of extracellular matrix and adhesion molecules between normal livers and cholestatic livers, we could find a number of potential molecules involved in liver regeneration. Our in vitro evaluations indicated that laminin alpha-3 significantly increased the number of liver cells (P<0.01 vs. Control) but decreased the proportion of claudin-3-positive hepatic stem cells (P<0.05 vs. Control). In contrast, thrombospondin-1 significantly reduced cell apoptosis (P<0.05 vs. Control) and maintained the proportion of claudin-3-positive hepatic stem cells. Otherwise, the combination of laminin alpha-3 and thrombospondin-1 increased the proliferation of liver cells. Based on our data, laminin alpha-3 and trombospondin-1 differently regulate the survival and differentiation of hepatocytes and hepatic stem cells, but relevant mechanisms are required to be elucidated by further study.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People’s HospitalGuangzhou 510180, China
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Heba E Sharaf Eldin
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University1-12-4 Sakamoto, Nagasaki 852-8523, Japan
- Department of Histology and Cell Biology, Faculty of Medicine, Tanta UniversityEgypt
| | - Wei-Li Gu
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People’s HospitalGuangzhou 510180, China
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
20
|
Rasouli M, Rahimi A, Soleimani M, keshel SH. The interplay between extracellular matrix and progenitor/stem cells during wound healing: Opportunities and future directions. Acta Histochem 2021; 123:151785. [PMID: 34500185 DOI: 10.1016/j.acthis.2021.151785] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/31/2021] [Accepted: 08/27/2021] [Indexed: 01/11/2023]
Abstract
Skin wound healing, a dynamic physiological process, progresses through coordinated overlapping phases to restore skin integrity. In some pathological conditions such as diabetes, wounds become chronic and hard-to-heal resulting in substantial morbidity and healthcare costs. Despite much advancement in understanding mechanisms of wound healing, chronic and intractable wounds are still a considerable challenge to nations' health care systems. Extracellular matrix (ECM) components play pivotal roles in all phases of wound healing. Therefore, a better understanding of their roles during wound healing can help improve wound care approaches. The ECM provides a 3D structure and forms the stem cell niche to support stem cell adhesion and survival and to regulate stem cell behavior and fate. Also, this dynamic structure reserves growth factors, regulates their bioavailability and provides biological signals. In various diseases, the composition and stiffness of the ECM is altered, which as a result, disrupts bidirectional cell-ECM interactions and tissue regeneration. Hence, due to the impact of ECM changes on stem cell fate during wound healing and the possibility of exploring new strategies to treat chronic wounds through manipulation of these interactions, in this review, we will discuss the importance/impact of ECM in the regulation of stem cell function and behavior to find ideal wound repair and regeneration strategies. We will also shed light on the necessity of using ECM in future wound therapy and highlight the potential roles of various biomimetic and ECM-based scaffolds as functional ECM preparations to mimic the native stem cell niche.
Collapse
|
21
|
Kim MS, Ha SE, Wu M, Zogg H, Ronkon CF, Lee MY, Ro S. Extracellular Matrix Biomarkers in Colorectal Cancer. Int J Mol Sci 2021; 22:9185. [PMID: 34502094 PMCID: PMC8430714 DOI: 10.3390/ijms22179185] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022] Open
Abstract
The cellular microenvironment composition and changes therein play an extremely important role in cancer development. Changes in the extracellular matrix (ECM), which constitutes a majority of the tumor stroma, significantly contribute to the development of the tumor microenvironment. These alterations within the ECM and formation of the tumor microenvironment ultimately lead to tumor development, invasion, and metastasis. The ECM is composed of various molecules such as collagen, elastin, laminin, fibronectin, and the MMPs that cleave these protein fibers and play a central role in tissue remodeling. When healthy cells undergo an insult like DNA damage and become cancerous, if the ECM does not support these neoplastic cells, further development, invasion, and metastasis fail to occur. Therefore, ECM-related cancer research is indispensable, and ECM components can be useful biomarkers as well as therapeutic targets. Colorectal cancer specifically, is also affected by the ECM and many studies have been conducted to unravel the complex association between the two. Here we summarize the importance of several ECM components in colorectal cancer as well as their potential roles as biomarkers.
Collapse
Affiliation(s)
- Min-Seob Kim
- Department of Physiology, Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.W.)
| | - Se-Eun Ha
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA; (S.-E.H.); (H.Z.); (C.F.R.)
| | - Moxin Wu
- Department of Physiology, Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.W.)
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang 332000, China
| | - Hannah Zogg
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA; (S.-E.H.); (H.Z.); (C.F.R.)
| | - Charles F. Ronkon
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA; (S.-E.H.); (H.Z.); (C.F.R.)
| | - Moon-Young Lee
- Department of Physiology, Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.W.)
| | - Seungil Ro
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA; (S.-E.H.); (H.Z.); (C.F.R.)
| |
Collapse
|
22
|
Chou K, Chang A, Ho C, Tsai T, Chen H, Chen P, Hwang TI. Thrombospondin-4 promotes bladder cancer cell migration and invasion via MMP2 production. J Cell Mol Med 2021; 25:6046-6055. [PMID: 34142438 PMCID: PMC8406484 DOI: 10.1111/jcmm.16463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 02/01/2021] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
Bladder cancer (BC) is the second most common urological tumour in Western countries. Approximately, 80% of patients with BC will present with non-muscle invasive bladder cancer (NMIBC), whereas a quarter will have muscle invasive disease (MIBC) at the time of BC diagnosis. However, patients with NMIBC are at risk of BC recurrence or progression into MIBC, and an MIBC prognosis is determined by the presence of progression and metastasis. Matrix metalloproteinase 2 (MMP2), a type of matrix metalloproteinase (MMP), plays a major role in tumour invasion and is well-characterized in BC prognosis. In BC, the mechanisms regulating MMP2 expression, and, in turn, promote cancer invasion, have hardly been explored. Thrombospondin-4 (THBS4/TSP4) is a matricellular glycoprotein that regulates multiple biological functions, including proliferation, angiogenesis, cell adhesion and extracellular matrix modelling. Based on the results of a meta-analysis in the Gene Expression Profiling Interactive Analysis 2 database, we observed that TSP4 expression levels were consistent with overall survival (OS) rate and BC progression, with the highest expression levels observed in the advanced stages of BC and associated with poor OS rate. In our pilot experiments, incubation with recombinant TSP4 promoted the migration and invasion in BC cells. Furthermore, MMP2 expression levels increased after recombinant TSP4 incubation. TSP4-induced-MMP2 expression and cell motility were regulated via the AKT signalling pathway. Our findings facilitate further investigation into TSP4 silencing-based therapeutic strategies for BC.
Collapse
Affiliation(s)
- Kuang‐Yu Chou
- Division of UrologyDepartment of SurgeryShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- Division of UrologySchool of MedicineFu‐Jen Catholic UniversityNew TaipeiTaiwan
| | - An‐Chen Chang
- Translational Medicine CenterShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
| | - Chao‐Yen Ho
- Division of UrologyDepartment of SurgeryShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- School of MedicineInstitute of Traditional MedicineNational Yang‐Ming UniversityTaipeiTaiwan
| | - Te‐Fu Tsai
- Division of UrologyDepartment of SurgeryShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- Division of UrologySchool of MedicineFu‐Jen Catholic UniversityNew TaipeiTaiwan
| | - Hung‐En Chen
- Division of UrologyDepartment of SurgeryShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
| | - Po‐Chun Chen
- Translational Medicine CenterShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- Department of BiotechnologyCollege of Health ScienceAsia UniversityTaichungTaiwan
- Department of Medical ResearchChina Medical University HospitalChina Medical UniversityTaichungTaiwan
| | - Thomas I‐Sheng Hwang
- Division of UrologyDepartment of SurgeryShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- Division of UrologySchool of MedicineFu‐Jen Catholic UniversityNew TaipeiTaiwan
- Department of UrologyTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
23
|
Thrombospondin-1 CD47 Signalling: From Mechanisms to Medicine. Int J Mol Sci 2021; 22:ijms22084062. [PMID: 33920030 PMCID: PMC8071034 DOI: 10.3390/ijms22084062] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/19/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Recent advances provide evidence that the cellular signalling pathway comprising the ligand-receptor duo of thrombospondin-1 (TSP1) and CD47 is involved in mediating a range of diseases affecting renal, vascular, and metabolic function, as well as cancer. In several instances, research has barely progressed past pre-clinical animal models of disease and early phase 1 clinical trials, while for cancers, anti-CD47 therapy has emerged from phase 2 clinical trials in humans as a crucial adjuvant therapeutic agent. This has important implications for interventions that seek to capitalize on targeting this pathway in diseases where TSP1 and/or CD47 play a role. Despite substantial progress made in our understanding of this pathway in malignant and cardiovascular disease, knowledge and translational gaps remain regarding the role of this pathway in kidney and metabolic diseases, limiting identification of putative drug targets and development of effective treatments. This review considers recent advances reported in the field of TSP1-CD47 signalling, focusing on several aspects including enzymatic production, receptor function, interacting partners, localization of signalling, matrix-cellular and cell-to-cell cross talk. The potential impact that these newly described mechanisms have on health, with a particular focus on renal and metabolic disease, is also discussed.
Collapse
|
24
|
Asparuhova MB, Stähli A, Guldener K, Sculean A. A Novel Volume-Stable Collagen Matrix Induces Changes in the Behavior of Primary Human Oral Fibroblasts, Periodontal Ligament, and Endothelial Cells. Int J Mol Sci 2021; 22:ijms22084051. [PMID: 33919968 PMCID: PMC8070954 DOI: 10.3390/ijms22084051] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study was to investigate the influence of a novel volume-stable collagen matrix (vCM) on early wound healing events including cellular migration and adhesion, protein adsorption and release, and the dynamics of the hemostatic system. For this purpose, we utilized transwell migration and crystal violet adhesion assays, ELISAs for quantification of adsorbed and released from the matrix growth factors, and qRT-PCR for quantification of gene expression in cells grown on the matrix. Our results demonstrated that primary human oral fibroblasts, periodontal ligament, and endothelial cells exhibited increased migration toward vCM compared to control cells that migrated in the absence of the matrix. Cellular adhesive properties on vCM were significantly increased compared to controls. Growth factors TGF-β1, PDGF-BB, FGF-2, and GDF-5 were adsorbed on vCM with great efficiency and continuously delivered in the medium after an initial burst release within hours. We observed statistically significant upregulation of genes encoding the antifibrinolytic thrombomodulin, plasminogen activator inhibitor type 1, thrombospondin 1, and thromboplastin, as well as strong downregulation of genes encoding the profibrinolytic tissue plasminogen activator, urokinase-type plasminogen activator, its receptor, and the matrix metalloproteinase 14 in cells grown on vCM. As a general trend, the stimulatory effect of the vCM on the expression of antifibrinolytic genes was synergistically enhanced by TGF-β1, PDGF-BB, or FGF-2, whereas the strong inhibitory effect of the vCM on the expression of profibrinolytic genes was reversed by PDGF-BB, FGF-2, or GDF-5. Taken together, our data strongly support the effect of the novel vCM on fibrin clot stabilization and coagulation/fibrinolysis equilibrium, thus facilitating progression to the next stages of the soft tissue healing process.
Collapse
Affiliation(s)
- Maria B. Asparuhova
- Dental Research Center, Laboratory of Oral Cell Biology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland; (A.S.); (K.G.); (A.S.)
- Correspondence:
| | - Alexandra Stähli
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland; (A.S.); (K.G.); (A.S.)
| | - Kevin Guldener
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland; (A.S.); (K.G.); (A.S.)
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland; (A.S.); (K.G.); (A.S.)
| |
Collapse
|
25
|
COMP and TSP-4: Functional Roles in Articular Cartilage and Relevance in Osteoarthritis. Int J Mol Sci 2021; 22:ijms22052242. [PMID: 33668140 PMCID: PMC7956748 DOI: 10.3390/ijms22052242] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a slow-progressing joint disease, leading to the degradation and remodeling of the cartilage extracellular matrix (ECM). The usually quiescent chondrocytes become reactivated and accumulate in cell clusters, become hypertrophic, and intensively produce not only degrading enzymes, but also ECM proteins, like the cartilage oligomeric matrix protein (COMP) and thrombospondin-4 (TSP-4). To date, the functional roles of these newly synthesized proteins in articular cartilage are still elusive. Therefore, we analyzed the involvement of both proteins in OA specific processes in in vitro studies, using porcine chondrocytes, isolated from femoral condyles. The effect of COMP and TSP-4 on chondrocyte migration was investigated in transwell assays and their potential to modulate the chondrocyte phenotype, protein synthesis and matrix formation by immunofluorescence staining and immunoblot. Our results demonstrate that COMP could attract chondrocytes and may contribute to a repopulation of damaged cartilage areas, while TSP-4 did not affect this process. In contrast, both proteins similarly promoted the synthesis and matrix formation of collagen II, IX, XII and proteoglycans, but inhibited that of collagen I and X, resulting in a stabilized chondrocyte phenotype. These data suggest that COMP and TSP-4 activate mechanisms to protect and repair the ECM in articular cartilage.
Collapse
|
26
|
Ganguly R, Khanal S, Mathias A, Gupta S, Lallo J, Sahu S, Ohanyan V, Patel A, Storm K, Datta S, Raman P. TSP-1 (Thrombospondin-1) Deficiency Protects ApoE -/- Mice Against Leptin-Induced Atherosclerosis. Arterioscler Thromb Vasc Biol 2021; 41:e112-e127. [PMID: 33327743 PMCID: PMC8105272 DOI: 10.1161/atvbaha.120.314962] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Hyperleptinemia, hallmark of obesity, is a putative pathophysiologic trigger for atherosclerosis. We previously reported a stimulatory effect of leptin on TSP-1 (thrombospondin-1) expression, a proatherogenic matricellular protein implicated in atherogenesis. However, a causal role of TSP-1 in leptin-driven atherosclerosis remains unknown. Approach and Results: Seventeen-weeks-old ApoE-/- and TSP-1-/-/ApoE-/- double knockout mice, on normocholesterolemic diet, were treated with or without murine recombinant leptin (5 µg/g bwt, IP) once daily for 3 weeks. Using aortic root morphometry and en face lesion assay, we found that TSP-1 deletion abrogated leptin-stimulated lipid-filled lesion burden, plaque area, and collagen accumulation in aortic roots of ApoE-/- mice, shown via Oil red O, hematoxylin and eosin, and Masson trichrome staining, respectively. Immunofluorescence microscopy of aortic roots showed that TSP-1 deficiency blocked leptin-induced inflammatory and smooth muscle cell abundance as well as cellular proliferation in ApoE-/- mice. Moreover, these effects were concomitant to changes in VLDL (very low-density lipoprotein)-triglyceride and HDL (high-density lipoprotein)-cholesterol levels. Immunoblotting further revealed reduced vimentin and pCREB (phospho-cyclic AMP response element-binding protein) accompanied with augmented smooth muscle-myosin heavy chain expression in aortic vessels of leptin-treated double knockout versus leptin-treated ApoE-/-; also confirmed in aortic smooth muscle cells from the mice genotypes, incubated ± leptin in vitro. Finally, TSP-1 deletion impeded plaque burden in leptin-treated ApoE-/- on western diet, independent of plasma lipid alterations. CONCLUSIONS The present study provides evidence for a protective effect of TSP-1 deletion on leptin-stimulated atherogenesis. Our findings suggest a regulatory role of TSP-1 on leptin-induced vascular smooth muscle cell phenotypic transition and inflammatory lesion invasion. Collectively, these results underscore TSP-1 as a potential target of leptin-induced vasculopathy.
Collapse
MESH Headings
- Animals
- Aorta/metabolism
- Aorta/pathology
- Aortic Diseases/chemically induced
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Atherosclerosis/chemically induced
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Cell Differentiation
- Cell Proliferation
- Cells, Cultured
- Collagen/metabolism
- Diet, High-Fat
- Disease Models, Animal
- Leptin
- Male
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Plaque, Atherosclerotic
- Signal Transduction
- Thrombospondin 1/deficiency
- Thrombospondin 1/genetics
- Mice
Collapse
Affiliation(s)
- Rituparna Ganguly
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
- School of Biomedical Sciences, Kent State University, Kent, OH
- Current Address: Department of Diabetes Complications and Metabolism, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Saugat Khanal
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
- School of Biomedical Sciences, Kent State University, Kent, OH
| | - Amy Mathias
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
| | - Shreya Gupta
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
- School of Biomedical Sciences, Kent State University, Kent, OH
| | - Jason Lallo
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
| | - Soumyadip Sahu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
- School of Biomedical Sciences, Kent State University, Kent, OH
- Current Address: National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
- School of Biomedical Sciences, Kent State University, Kent, OH
| | - Aakaash Patel
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
| | - Kyle Storm
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
| | - Sujay Datta
- Department of Statistics, The University of Akron, Akron, OH
| | - Priya Raman
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
- School of Biomedical Sciences, Kent State University, Kent, OH
| |
Collapse
|
27
|
Zhang J, Wang H, Lv C, Han J, Hao M, Li J, Qiao H. Cartilage oligomeric matrix protein affects the biological behavior of papillary thyroid carcinoma cells by activating the PI3K/AKT/Bcl-2 pathway. J Cancer 2021; 12:1623-1633. [PMID: 33613749 PMCID: PMC7890313 DOI: 10.7150/jca.49144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 12/14/2020] [Indexed: 11/24/2022] Open
Abstract
Objective: To explore the effect of cartilage oligomeric matrix protein (COMP) on papillary thyroid carcinoma (PTC). Methods: COMP expression levels in PTC tissues and matched adjacent normal tissues were measured using tissue microarrays. Human PTC cells were cultured and transduced with lentiviral short hairpin RNA against COMP (COMP-shRNA), a negative control (NC) shRNA, or mock transfected (Control). We used the Cell Counting Kit-8, performed colony formation assays, wound healing assays, Transwell invasion assays, flow cytometry, and measured the expression of apoptosis-related proteins at the mRNA and protein levels to explore the effects of COMP on the biological behavior of PTC cells and to discover the specific signaling pathway involved in these processes. Results: COMP expression was significantly higher in PTC tissues than in adjacent normal tissues. At the cellular level, COMP promoted cell migration, increased the invasiveness of PTC cells, and inhibited apoptosis. However, differences in cell proliferation were only observed within 72 hours. At the same time, colony formation assays showed that silencing COMP inhibited the proliferation of PTC cells. We also found that COMP regulated the behavior of PTC cells by activating the PI3K/AKT/Bcl-2 pathway. Conclusions: COMP is upregulated in PTC, which enhances cancer cell invasion and inhibits apoptosis, contributing to the development and progression of PTC. Thus, COMP may serve as a new biomarker for PTC.
Collapse
Affiliation(s)
- Jirong Zhang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Hongzhi Wang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Chunpeng Lv
- Epidemiology and Health Statistics, Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Jun Han
- Department of Endocrine and Metabolism, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Mingyu Hao
- Department of Endocrine and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Jingjing Li
- Department of Endocrine and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Hong Qiao
- Department of Endocrine and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| |
Collapse
|
28
|
Pennarossa G, Arcuri S, De Iorio T, Gandolfi F, Brevini TAL. Current Advances in 3D Tissue and Organ Reconstruction. Int J Mol Sci 2021; 22:E830. [PMID: 33467648 PMCID: PMC7830719 DOI: 10.3390/ijms22020830] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Bi-dimensional culture systems have represented the most used method to study cell biology outside the body for over a century. Although they convey useful information, such systems may lose tissue-specific architecture, biomechanical effectors, and biochemical cues deriving from the native extracellular matrix, with significant alterations in several cellular functions and processes. Notably, the introduction of three-dimensional (3D) platforms that are able to re-create in vitro the structures of the native tissue, have overcome some of these issues, since they better mimic the in vivo milieu and reduce the gap between the cell culture ambient and the tissue environment. 3D culture systems are currently used in a broad range of studies, from cancer and stem cell biology, to drug testing and discovery. Here, we describe the mechanisms used by cells to perceive and respond to biomechanical cues and the main signaling pathways involved. We provide an overall perspective of the most recent 3D technologies. Given the breadth of the subject, we concentrate on the use of hydrogels, bioreactors, 3D printing and bioprinting, nanofiber-based scaffolds, and preparation of a decellularized bio-matrix. In addition, we report the possibility to combine the use of 3D cultures with functionalized nanoparticles to obtain highly predictive in vitro models for use in the nanomedicine field.
Collapse
Affiliation(s)
- Georgia Pennarossa
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Sharon Arcuri
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Teresina De Iorio
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy;
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| |
Collapse
|
29
|
Microglia-organized scar-free spinal cord repair in neonatal mice. Nature 2020; 587:613-618. [PMID: 33029008 PMCID: PMC7704837 DOI: 10.1038/s41586-020-2795-6] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
It is thought that spinal cord injury triggers scar formation with little axon regeneration in mammals1–4. Here we report that in neonatal mice, a crush injury to the spinal cord leads to a scar-free healing that permits the growth of long projecting axons through the lesion. Depletion of microglia in neonates disrupts such healing and stalls axon regrowth, suggesting a critical role for microglia in orchestrating the injury response. Using single cell RNA-sequencing and functional analyses, we discovered that neonatal microglia undergo a transient activation and play at least two critical roles in scar-free healing. First, they transiently secrete fibronectin and its binding proteins, to form extracellular matrix bridges that ligate the severed ends. Second, neonatal, but not adult, microglia express a number of extracellular and intracellular peptidase inhibitors, along with other molecules involved in inflammatory resolution. Strikingly, upon transplantation into adult spinal cord lesions, both adult microglia treated with peptidases inhibitors and neonatal microglia significantly improve healing and axon regrowth. Together, our results reveal the cellular and molecular basis underlying the nearly complete recovery after spinal cord injury in neonatal mice, pointing to potential strategies to facilitate scar-free healing in the adult mammalian nervous system.
Collapse
|
30
|
Dreiner M, Willwacher S, Kramer A, Kümmel J, Frett T, Zaucke F, Liphardt AM, Gruber M, Niehoff A. Short-term Response of Serum Cartilage Oligomeric Matrix Protein to Different Types of Impact Loading Under Normal and Artificial Gravity. Front Physiol 2020; 11:1032. [PMID: 32982779 PMCID: PMC7489036 DOI: 10.3389/fphys.2020.01032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/28/2020] [Indexed: 11/13/2022] Open
Abstract
Microgravity during long-term space flights induces degeneration of articular cartilage. Artificial gravity through centrifugation combined with exercise has been suggested as a potential countermeasure for musculoskeletal degeneration. The purpose of this study was to investigate the effect of different types of impact loading under normal and artificial gravity conditions on serum concentrations of cartilage oligomeric matrix protein (COMP), a biomarker of cartilage metabolism. Fifteen healthy male adults (26 ± 4 years, 181 ± 4 cm, 77 ± 6 kg) performed four different 30-min impact loading protocols on four experimental days: jumping with artificial gravity elicited by centrifugation in a short-arm centrifuge (AGJ), jumping with artificial gravity generated by low-pressure cylinders in a sledge jump system (SJS), vertical jumping under Earth gravity (EGJ), and running under Earth gravity (RUN). Five blood samples per protocol were taken: 30 min before, immediately before, immediately after, 30 min after, and 60 min after impact loading. Serum COMP concentrations were analyzed in these samples. During the impact exercises, ground reaction forces were recorded. Peak ground reaction forces were significantly different between the three jumping protocols (p < 0.001), increasing from AGJ (14 N/kg) to SJS (22 N/kg) to EGJ (29 N/kg) but were similar in RUN (22 N/kg) compared to SJS. The serum COMP concentration was increased (p < 0.001) immediately after all loading protocols, and then decreased (p < 0.001) at 30 min post-exercise compared to immediately after the exercise. Jumping and running under Earth gravity (EGJ and RUN) resulted in a significantly higher (p < 0.05) increase of serum COMP levels 30 min after impact loading compared to the impact loading under artificial gravity (RUN +30%, EGJ +20%, AGJ +17%, and SJS +13% compared to baseline). In conclusion, both the amplitude and the number of the impacts contribute to inducing higher COMP responses and are therefore likely important factors affecting cartilage metabolism. RUN had the largest effect on serum COMP concentration, presumably due to the high number of impacts, which was 10 times higher than for the jump modalities. Future studies should aim at establishing a dose-response relationship for different types of exercise using comparable amounts of impacts.
Collapse
Affiliation(s)
- Maren Dreiner
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Cologne, Germany
| | - Steffen Willwacher
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Cologne, Germany
| | - Andreas Kramer
- Human Performance Research Centre, Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Jakob Kümmel
- Human Performance Research Centre, Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Timo Frett
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim gGmbH, Frankfurt/Main, Germany
| | - Anna-Maria Liphardt
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Cologne, Germany.,Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Markus Gruber
- Human Performance Research Centre, Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Faculty of Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
31
|
Carminati L, Taraboletti G. Thrombospondins in bone remodeling and metastatic bone disease. Am J Physiol Cell Physiol 2020; 319:C980-C990. [PMID: 32936697 DOI: 10.1152/ajpcell.00383.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Thrombospondins (TSPs) are a family of five multimeric matricellular proteins. Through a wide range of interactions, TSPs play pleiotropic roles in embryogenesis and in tissue remodeling in adult physiology as well as in pathological conditions, including cancer development and metastasis. TSPs are active in bone remodeling, the process of bone resorption (osteolysis) and deposition (osteogenesis) that maintains bone homeostasis. TSPs are particularly involved in aberrant bone remodeling, including osteolytic and osteoblastic skeletal cancer metastasis, frequent in advanced cancers such as breast and prostate carcinoma. TSPs are major players in the bone metastasis microenvironment, where they finely tune the cross talk between tumor cells and bone resident cells in the metastatic niche. Each TSP family member has different effects on the differentiation and activity of bone cells-including the bone-degrading osteoclasts and the bone-forming osteoblasts-with different outcomes on the development and growth of osteolytic and osteoblastic metastases. Here, we overview the involvement of TSP family members in the bone tissue microenvironment, focusing on their activity on osteoclasts and osteoblasts in bone remodeling, and present the evidence to date of their roles in bone metastasis establishment and growth.
Collapse
Affiliation(s)
- Laura Carminati
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giulia Taraboletti
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
32
|
Arun A, Rayford KJ, Cooley A, Rachakonda G, Villalta F, Pratap S, Lima MF, Sheibani N, Nde PN. Thrombospondin-1 Plays an Essential Role in Yes-Associated Protein Nuclear Translocation during the Early Phase of Trypanosoma cruzi Infection in Heart Endothelial Cells. Int J Mol Sci 2020; 21:ijms21144912. [PMID: 32664627 PMCID: PMC7403984 DOI: 10.3390/ijms21144912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 01/03/2023] Open
Abstract
The protozoan parasite Trypanosoma cruzi is the causative agent of Chagas disease. This neglected tropical disease causes severe morbidity and mortality in endemic regions. About 30% of T. cruzi infected individuals will present with cardiac complications. Invasive trypomastigotes released from infected cells can be carried in the vascular endothelial system to infect neighboring and distant cells. During the process of cellular infection, the parasite induces host cells, to increase the levels of host thrombospondin-1 (TSP-1), to facilitate the process of infection. TSP-1 plays important roles in the functioning of vascular cells, including vascular endothelial cells with important implications in cardiovascular health. Many signal transduction pathways, including the yes-associated protein 1 (YAP)/transcriptional coactivator, with PDZ-binding motif (TAZ) signaling, which are upstream of TSP-1, have been linked to the pathophysiology of heart damage. The molecular mechanisms by which T. cruzi signals, and eventually infects, heart endothelial cells remain unknown. To evaluate the importance of TSP-1 expression in heart endothelial cells during the process of T. cruzi infection, we exposed heart endothelial cells prepared from Wild Type and TSP-1 Knockout mouse to invasive T. cruzi trypomastigotes at multiple time points, and evaluated changes in the hippo signaling cascade using immunoblotting and immunofluorescence assays. We found that the parasite turned off the hippo signaling pathway in TSP-1KO heart endothelial cells. The levels of SAV1 and MOB1A increased to a maximum of 2.70 ± 0.23 and 5.74 ± 1.45-fold at 3 and 6 h, respectively, in TSP-1KO mouse heart endothelial cells (MHEC), compared to WT MHEC, following a parasite challenge. This was accompanied by a significant continuous increase in the nuclear translocation of downstream effector molecule YAP, to a maximum mean nuclear fluorescence intensity of 10.14 ± 0.40 at 6 h, compared to wild type cells. Furthermore, we found that increased nuclear translocated YAP significantly colocalized with the transcription co-activator molecule pan-TEAD, with a maximum Pearson's correlation coefficient of 0.51 ± 0.06 at 6 h, compared to YAP-Pan-TEAD colocalization in the WT MHEC, which decreased significantly, with a minimum Pearson's correlation coefficient of 0.30 ± 0.01 at 6 h. Our data indicate that, during the early phase of infection, upregulated TSP-1 is essential for the regulation of the hippo signaling pathway. These studies advance our understanding of the molecular interactions occurring between heart endothelial cells and T. cruzi, in the presence and absence of TSP-1, providing insights into processes linked to parasite dissemination and pathogenesis.
Collapse
Affiliation(s)
- Ashutosh Arun
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Ayorinde Cooley
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Girish Rachakonda
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Fernando Villalta
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Siddharth Pratap
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (S.P.); (M.F.L.)
| | - Maria F. Lima
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (S.P.); (M.F.L.)
- Department of Molecular Cellular and Biomedical Sciences, School of Medicine, The City College of New York, New York, NY 10031, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
- Correspondence: ; Tel.: +1-615-327-6997
| |
Collapse
|
33
|
Shihan MH, Kanwar M, Wang Y, Jackson EE, Faranda AP, Duncan MK. Fibronectin has multifunctional roles in posterior capsular opacification (PCO). Matrix Biol 2020; 90:79-108. [PMID: 32173580 DOI: 10.1016/j.matbio.2020.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/10/2020] [Accepted: 02/25/2020] [Indexed: 12/26/2022]
Abstract
Fibrotic posterior capsular opacification (PCO), one of the major complications of cataract surgery, occurs when lens epithelial cells (LCs) left behind post cataract surgery (PCS) undergo epithelial to mesenchymal transition, migrate into the optical axis and produce opaque scar tissue. LCs left behind PCS robustly produce fibronectin, although its roles in fibrotic PCO are not known. In order to determine the function of fibronectin in PCO pathogenesis, we created mice lacking the fibronectin gene (FN conditional knock out -FNcKO) from the lens. While animals from this line have normal lenses, upon lens fiber cell removal which models cataract surgery, FNcKO LCs exhibit a greatly attenuated fibrotic response from 3 days PCS onward as assessed by a reduction in surgery-induced cell proliferation, and fibrotic extracellular matrix (ECM) production and deposition. This is correlated with less upregulation of Transforming Growth Factor β (TGFβ) and integrin signaling in FNcKO LCs PCS concomitant with sustained Bone Morphogenetic Protein (BMP) signaling and elevation of the epithelial cell marker E cadherin. Although the initial fibrotic response of FNcKO LCs was qualitatively normal at 48 h PCS as measured by the upregulation of fibrotic marker protein αSMA, RNA sequencing revealed that the fibrotic response was already quantitatively attenuated at this time, as measured by the upregulation of mRNAs encoding molecules that control, and are controlled by, TGFβ signaling, including many known markers of fibrosis. Most notably, gremlin-1, a known regulator of TGFβ superfamily signaling, was upregulated sharply in WT LCs PCS, while this response was attenuated in FNcKO LCs. As exogenous administration of either active TGFβ1 or gremlin-1 to FNcKO lens capsular bags rescued the attenuated fibrotic response of fibronectin null LCs PCS including the loss of SMAD2/3 phosphorylation, this suggests that fibronectin plays multifunctional roles in fibrotic PCO development.
Collapse
Affiliation(s)
- Mahbubul H Shihan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Mallika Kanwar
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Yan Wang
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Erin E Jackson
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Adam P Faranda
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
34
|
Bozó R, Szél E, Danis J, Gubán B, Bata-Csörgő Z, Szabó K, Kemény L, Groma G. Cartilage Oligomeric Matrix Protein Negatively Influences Keratinocyte Proliferation via α5β1-Integrin: Potential Relevance of Altered Cartilage Oligomeric Matrix Protein Expression in Psoriasis. J Invest Dermatol 2020; 140:1733-1742.e7. [PMID: 32057837 DOI: 10.1016/j.jid.2019.12.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 11/27/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023]
Abstract
In psoriasis, nonlesional skin shows alterations at the dermal-epidermal junction compared with healthy skin. Cartilage oligomeric matrix protein (COMP) is part of the papillary dermis of healthy skin, and its expression has not yet been studied in psoriatic skin. In this study, we found that COMP localization extended deeper into the dermis and formed a more continuous layer in psoriatic nonlesional skin compared with healthy skin, whereas in psoriatic lesions, COMP showed a partially discontinuous deposition at the dermal-epidermal junction. COMP and β1-integrin showed strong colocalization in nonlesional skin, where the laminin layer within the basement membrane is discontinuous. In in vitro models, the presence of exogenous COMP decreased the proliferation rate of keratinocytes, and this proliferation-suppressing effect was diminished by blocking α5β1-integrin. Our results suggest that COMP can interact with α5β1-integrin of basal keratinocytes through the disrupted basement membrane, and this interaction might stabilize the epidermis in the nonlesional state by contributing to the suppression of keratinocyte proliferation. The antiproliferative effect of COMP is likely to be relevant to other skin diseases in which chronic nonhealing wounds are coupled with massive COMP accumulation.
Collapse
Affiliation(s)
- Renáta Bozó
- Department of Dermatology and Allergology University of Szeged, Szeged, Hungary
| | - Edit Szél
- Department of Dermatology and Allergology University of Szeged, Szeged, Hungary
| | - Judit Danis
- Department of Dermatology and Allergology University of Szeged, Szeged, Hungary; MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Barbara Gubán
- Department of Dermatology and Allergology University of Szeged, Szeged, Hungary
| | - Zsuzsanna Bata-Csörgő
- Department of Dermatology and Allergology University of Szeged, Szeged, Hungary; MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Kornélia Szabó
- Department of Dermatology and Allergology University of Szeged, Szeged, Hungary; MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology University of Szeged, Szeged, Hungary; MTA-SZTE Dermatological Research Group, Szeged, Hungary; HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Gergely Groma
- Department of Dermatology and Allergology University of Szeged, Szeged, Hungary; MTA-SZTE Dermatological Research Group, Szeged, Hungary.
| |
Collapse
|
35
|
Sui Z, Sun H, Weng Y, Zhang X, Sun M, Sun R, Zhao B, Liang Z, Zhang Y, Li C, Zhang L. Quantitative proteomics analysis of deer antlerogenic periosteal cells reveals potential bioactive factors in velvet antlers. J Chromatogr A 2020; 1609:460496. [DOI: 10.1016/j.chroma.2019.460496] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/17/2019] [Accepted: 08/28/2019] [Indexed: 01/15/2023]
|
36
|
Pandey R, Zhou M, Islam S, Chen B, Barker NK, Langlais P, Srivastava A, Luo M, Cooke LS, Weterings E, Mahadevan D. Carcinoembryonic antigen cell adhesion molecule 6 (CEACAM6) in Pancreatic Ductal Adenocarcinoma (PDA): An integrative analysis of a novel therapeutic target. Sci Rep 2019; 9:18347. [PMID: 31797958 PMCID: PMC6893022 DOI: 10.1038/s41598-019-54545-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
We investigated biomarker CEACAM6, a highly abundant cell surface adhesion receptor that modulates the extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDA). The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) RNA-Seq data from PDA patients were analyzed for CEACAM6 expression and evaluated for overall survival, association, enrichment and correlations. A CRISPR/Cas9 Knockout (KO) of CEACAM6 in PDA cell line for quantitative proteomics, mitochondrial bioenergetics and tumor growth in mice were conducted. We found CEACAM6 is over-expressed in primary and metastatic basal and classical PDA subtypes. Highest levels are in classical activated stroma subtype. CEACAM6 over-expression is universally a poor prognostic marker in KRAS mutant and wild type PDA. High CEACAM6 expression is associated with low cytolytic T-cell activity in both basal and classical PDA subtypes and correlates with low levels of T-REG markers. In HPAF-II cells knockout of CEACAM6 alters ECM-cell adhesion, catabolism, immune environment, transmembrane transport and autophagy. CEACAM6 loss increases mitochondrial basal and maximal respiratory capacity. HPAF-II CEACAM6−/− cells are growth suppressed by >65% vs. wild type in mice bearing tumors. CEACAM6, a key regulator affects several hallmarks of PDA including the fibrotic reaction, immune regulation, energy metabolism and is a novel therapeutic target in PDA.
Collapse
Affiliation(s)
- Ritu Pandey
- University of Arizona Cancer Center, University of Arizona, Tucson, USA. .,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, USA.
| | - Muhan Zhou
- University of Arizona Cancer Center, University of Arizona, Tucson, USA
| | - Shariful Islam
- University of Arizona Cancer Center, University of Arizona, Tucson, USA
| | - Baowei Chen
- University of Arizona Cancer Center, University of Arizona, Tucson, USA
| | - Natalie K Barker
- Department of Medicine, College of Medicine, University of Arizona, Tucson, USA
| | - Paul Langlais
- Department of Medicine, College of Medicine, University of Arizona, Tucson, USA
| | - Anup Srivastava
- Department of Medicine, College of Medicine, University of Arizona, Tucson, USA
| | - Moulun Luo
- Department of Medicine, College of Medicine, University of Arizona, Tucson, USA
| | - Laurence S Cooke
- University of Arizona Cancer Center, University of Arizona, Tucson, USA
| | - Eric Weterings
- University of Arizona Cancer Center, University of Arizona, Tucson, USA.,Department of Medicine, College of Medicine, University of Arizona, Tucson, USA.,Department of Radiation Oncology, College of Medicine, University of Arizona, Tucson, USA
| | - Daruka Mahadevan
- University of Arizona Cancer Center, University of Arizona, Tucson, USA. .,Department of Medicine, College of Medicine, University of Arizona, Tucson, USA.
| |
Collapse
|
37
|
Mammoto A, Mammoto T. Vascular Niche in Lung Alveolar Development, Homeostasis, and Regeneration. Front Bioeng Biotechnol 2019; 7:318. [PMID: 31781555 PMCID: PMC6861452 DOI: 10.3389/fbioe.2019.00318] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/25/2019] [Indexed: 12/28/2022] Open
Abstract
Endothelial cells (ECs) constitute small capillary blood vessels and contribute to delivery of nutrients, oxygen and cellular components to the local tissues, as well as to removal of carbon dioxide and waste products from the tissues. Besides these fundamental functions, accumulating evidence indicates that capillary ECs form the vascular niche. In the vascular niche, ECs reciprocally crosstalk with resident cells such as epithelial cells, mesenchymal cells, and immune cells to regulate development, homeostasis, and regeneration in various organs. Capillary ECs supply paracrine factors, called angiocrine factors, to the adjacent cells in the niche and orchestrate these processes. Although the vascular niche is anatomically and functionally well-characterized in several organs such as bone marrow and neurons, the effects of endothelial signals on other resident cells and anatomy of the vascular niche in the lung have not been well-explored. This review discusses the role of alveolar capillary ECs in the vascular niche during development, homeostasis and regeneration.
Collapse
Affiliation(s)
- Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tadanori Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
38
|
Hoesl C, Fröhlich T, Hundt JE, Kneitz H, Goebeler M, Wolf R, Schneider MR, Dahlhoff M. The transmembrane protein LRIG2 increases tumor progression in skin carcinogenesis. Mol Oncol 2019; 13:2476-2492. [PMID: 31580518 PMCID: PMC6822252 DOI: 10.1002/1878-0261.12579] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/18/2019] [Accepted: 10/01/2019] [Indexed: 01/23/2023] Open
Abstract
Over the last few decades, the number of cases of non‐melanoma skin cancer (NMSC) has risen to over 3 million cases every year worldwide. Members of the ERBB receptor family are important regulators of skin development and homeostasis and, when dysregulated, contribute to skin pathogenesis. In this study, we investigated leucine‐rich repeats and immunoglobulin‐like domains 2 (LRIG2), a transmembrane protein involved in feedback loop regulation of the ERBB receptor family during NMSC. LRIG2 was identified to be up‐regulated in various types of squamous cell carcinoma (SCC), but little is known about LRIG2 in cutaneous SCC (cSCC). To investigate the function of LRIG2 in cSCC in vivo, we generated a skin‐specific LRIG2 overexpressing transgenic mouse line (LRIG2‐TG) using the Tet‐Off system. We employed the 7,12‐dimethylbenz(a)anthracene/12‐O‐tetra‐decanoylphorbol‐13‐acetate (DMBA/TPA) two‐stage chemical carcinogenesis model and analyzed the skin during homeostasis and tumorigenesis. LRIG2‐TG mice did not exhibit alterations in skin development or homeostasis but showed an interaction between LRIG2 and thrombospondin‐1, which is often involved in angiogenesis and tumorigenesis. However, during carcinogenesis, transgenic animals showed significantly increased tumor progression and a more rapid development of cSCC. This was accompanied by changes in the ERBB system. After a single TPA application, inflammation of the epidermis was enhanced during LRIG2 overexpression. In human skin samples, LRIG2 expression was identified in the basal layer of the epidermis and in hair follicles of normal skin, but also in cSCC samples. In conclusion, epidermal LRIG2 excess is associated with activated EGFR/ERBB4‐MAPK signaling and accelerated tumor progression in experimentally induced NMSC, suggesting LRIG2 as a potential oncoprotein in skin.
Collapse
Affiliation(s)
- Christine Hoesl
- Institute of Molecular Animal Breeding and BiotechnologyGene CenterLMU MünchenGermany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA)Gene CenterLMU MünchenGermany
| | - Jennifer E. Hundt
- Lübeck Institute for Experimental DermatologyUniversität zu LübeckGermany
| | - Hermann Kneitz
- Klinik und Poliklinik für Dermatologie, Venerologie und AllergologieUniversitätsklinikum WürzburgGermany
| | - Matthias Goebeler
- Klinik und Poliklinik für Dermatologie, Venerologie und AllergologieUniversitätsklinikum WürzburgGermany
| | - Ronald Wolf
- Department of Dermatology und AllergologyPhilipps UniversityMarburgGermany
| | - Marlon R. Schneider
- Institute of Molecular Animal Breeding and BiotechnologyGene CenterLMU MünchenGermany
| | - Maik Dahlhoff
- Institute of Molecular Animal Breeding and BiotechnologyGene CenterLMU MünchenGermany
| |
Collapse
|
39
|
Kennedy SM, Sheridan C, Kearns VR, Bilir EK, Fan X, Grierson I, Choudhary A. Thrombospondin-2 is up-regulated by TGFβ2 and increases fibronectin expression in human trabecular meshwork cells. Exp Eye Res 2019; 189:107820. [PMID: 31589839 DOI: 10.1016/j.exer.2019.107820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 09/18/2019] [Accepted: 09/27/2019] [Indexed: 11/25/2022]
Abstract
Elevated intraocular pressure (IOP) is a major risk factor for the development of primary open-angle glaucoma (POAG). This is from an increased aqueous humour (AH) outflow resistance through the trabecular meshwork (TM). The pathogenic mechanisms leading to the increase in TM outflow resistance are poorly understood but are thought to be from a dysregulation of the TM extracellular matrix (ECM) environment. ECM modification and turnover are crucial in regulating the resistance to aqueous outflow. ECM turnover is influenced by a complex interplay of growth factors such as transforming growth factors (TGFβ) family and matrix metalloproteinases (MMPs). Elevated TGFβ2 levels result in an increase in ECM deposition such as fibronectin leading to increased resistance. Fibronectin is a major component of TM ECM and plays a key role in its maintenance. Thrombospondins (TSP)-1 and -2 are important regulators of the ECM environment. TSP-1 has been implicated in the pathogenesis of POAG through activation of TGFβ2 within the TM. TSP-2 does not contain the catalytic domain to activate latent TGFβ, but is able to mediate the activities of MMP 2 and 9, thereby influencing ECM turnover. TSP-2 knock out mice show lower IOP levels compared to their wild type counterparts, suggesting the involvement of TSP-2 in the pathogenesis of POAG but its role in the pathogenesis of POAG remains unclear. The purpose of this study was to investigate the role of TSP-2 in trabecular meshwork ECM regulation and hence the pathogenesis of POAG. TSP-1 and TSP-2 expressions in immortalised glaucomatous TM cells (GTM3) and primary human non-glaucomatous (NTM) and glaucomatous cells (GTM) were determined by immunocytochemistry, immuno-blot analysis and qPCR following treatment with TGFβ2 and Dexamethasone. The level of ECM protein fibronectin was determined in TM cells using immuno-blot analysis following treatment with TSP-1 or -2. TM cells secrete TSP-1 and -2 under basal conditions at the protein level and TSP-2 mRNA and protein levels were increased in response to TGFβ2 three days post treatment. Exogenous treatment with TSP-2 up-regulated the expression of fibronectin protein in GTM3 cells, primary NTM and GTM cells. TSP-1 did not affect fibronectin protein levels in GTM3 cells. This suggests that the role of TSP-2 might be distinct from that of TSP-1 in the regulation of the TM cell ECM environment. TSP-2 may be involved in the pathogenesis of POAG and contribute to increased IOP levels by increasing the deposition of fibronectin within the ECM in response to TGFβ2.
Collapse
Affiliation(s)
- Stephnie Michelle Kennedy
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 west Derby Street, Liverpool, L69 8TX, UK.
| | - Carl Sheridan
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 west Derby Street, Liverpool, L69 8TX, UK.
| | - Victoria Rosalind Kearns
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 west Derby Street, Liverpool, L69 8TX, UK.
| | - Emine Kubra Bilir
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 west Derby Street, Liverpool, L69 8TX, UK.
| | - Xiaochen Fan
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 west Derby Street, Liverpool, L69 8TX, UK.
| | - Ian Grierson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 west Derby Street, Liverpool, L69 8TX, UK.
| | - Anshoo Choudhary
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 west Derby Street, Liverpool, L69 8TX, UK; St Paul's Eye Unit, Royal Liverpool University Hospital, Liverpool, L7 8XP, UK.
| |
Collapse
|
40
|
Hwang HJ, Oh MS, Lee DW, Kuh HJ. Multiplex quantitative analysis of stroma-mediated cancer cell invasion, matrix remodeling, and drug response in a 3D co-culture model of pancreatic tumor spheroids and stellate cells. J Exp Clin Cancer Res 2019; 38:258. [PMID: 31200779 PMCID: PMC6567511 DOI: 10.1186/s13046-019-1225-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 05/10/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a stroma-rich carcinoma, and pancreatic stellate cells (PSCs) are a major component of this dense stroma. PSCs play significant roles in metastatic progression and chemoresistance through cross-talk with cancer cells. Preclinical in vitro tumor model of invasive phenotype should incorporate three-dimensional (3D) culture of cancer cells and PSCs in extracellular matrix (ECM) for clinical relevance and predictability. METHODS PANC-1 cells were cultured as tumor spheroids (TSs) using our previously developed minipillar chips, and co-cultured with PSCs, both embedded in collagen gels. Effects of PSC co-culture on ECM fiber network, invasive migration of cancer cells, and expression of epithelial-mesenchymal transition (EMT)-related proteins were examined. Conditioned media was also analyzed for secreted factors involved in cancer cell-PSC interactions. Inhibitory effect on cancer cell invasion was compared between gemcitabine and paclitaxel at an equitoxic concentration in PANC-1 TSs co-cultured with PSCs. RESULTS Co-culture condition was optimized for the growth of TSs, activation of PSCs, and their interaction. Increase in cancer cell invasion via ECM remodeling, invadopodia formation and EMT, as well as drug resistance was recapitulated in the TS-PSC co-culture, and appeared to be mediated by cancer cell-PSC interaction via multiple secreted factors, including IL-6, IL-8, IGF-1, EGF, TIMP-1, uPA, PAI-1, and TSP-1. Compared to gemcitabine, paclitaxel showed a greater anti-invasive activity, which was attributed to suppresion of invadopodia formation in cancer cells as well as to PSC-specific cytotoxicity abrogating its paracrine signaling. CONCLUSIONS Here, we established 3D co-culture of TSs of PANC-1 cells and PSCs using minipillar histochips as a novel tumoroid model of PDAC. Our results indicate usefulness of the present co-culture model and multiplex quantitative analysis method not only in studying the role of PSCs and their interactions with tumor cell towards metastatic progression, but also in the drug evaluation of stroma-targeting drugs.
Collapse
Affiliation(s)
- Hyun Ju Hwang
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min-Suk Oh
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Woo Lee
- Departments of Biomedical Engineering, Konyang University, Daejeon, Republic of Korea
- Medical & Bio Device, #B-9, 145 Gwanggyo-ro, Suwon, Republic of Korea
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-ku, Seoul, 06591 Republic of Korea
| |
Collapse
|
41
|
Mostovenko E, Young T, Muldoon PP, Bishop L, Canal CG, Vucetic A, Zeidler-Erdely PC, Erdely A, Campen MJ, Ottens AK. Nanoparticle exposure driven circulating bioactive peptidome causes systemic inflammation and vascular dysfunction. Part Fibre Toxicol 2019; 16:20. [PMID: 31142334 PMCID: PMC6542040 DOI: 10.1186/s12989-019-0304-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background The mechanisms driving systemic effects consequent pulmonary nanoparticle exposure remain unclear. Recent work has established the existence of an indirect process by which factors released from the lung into the circulation promote systemic inflammation and cellular dysfunction, particularly on the vasculature. However, the composition of circulating contributing factors and how they are produced remains unknown. Evidence suggests matrix protease involvement; thus, here we used a well-characterized multi-walled carbon nanotube (MWCNT) oropharyngeal aspiration model with known vascular effects to assess the distinct contribution of nanoparticle-induced peptide fragments in driving systemic pathobiology. Results Data-independent mass spectrometry enabled the unbiased quantitative characterization of 841 significant MWCNT-responses within an enriched peptide fraction, with 567 of these factors demonstrating significant correlation across animal-paired bronchoalveolar lavage and serum biofluids. A database search curated for known matrix protease substrates and predicted signaling motifs enabled identification of 73 MWCNT-responsive peptides, which were significantly associated with an abnormal cardiovascular phenotype, extracellular matrix organization, immune-inflammatory processes, cell receptor signaling, and a MWCNT-altered serum exosome population. Production of a diverse peptidomic response was supported by a wide number of upregulated matrix and lysosomal proteases in the lung after MWCNT exposure. The peptide fraction was then found bioactive, producing endothelial cell inflammation and vascular dysfunction ex vivo akin to that induced with whole serum. Results implicate receptor ligand functionality in driving systemic effects, exemplified by an identified 59-mer thrombospondin fragment, replete with CD36 modulatory motifs, that when synthesized produced an anti-angiogenic response in vitro matching that of the peptide fraction. Other identified peptides point to integrin ligand functionality and more broadly to a diversity of receptor-mediated bioactivity induced by the peptidomic response to nanoparticle exposure. Conclusion The present study demonstrates that pulmonary-sequestered nanoparticles, such as multi-walled carbon nanotubes, acutely upregulate a diverse profile of matrix proteases, and induce a complex peptidomic response across lung and blood compartments. The serum peptide fraction, having cell-surface receptor ligand properties, conveys peripheral bioactivity in promoting endothelial cell inflammation, vasodilatory dysfunction and inhibiting angiogenesis. Results here establish peptide fragments as indirect, non-cytokine mediators and putative biomarkers of systemic health outcomes from nanoparticle exposure.
Collapse
Affiliation(s)
- Ekaterina Mostovenko
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Tamara Young
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Pretal P Muldoon
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Lindsey Bishop
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Christopher G Canal
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Aleksandar Vucetic
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Patti C Zeidler-Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA.
| |
Collapse
|
42
|
Kim T, Ahmad K, Shaikh S, Jan AT, Seo MG, Lee EJ, Choi I. Dermatopontin in Skeletal Muscle Extracellular Matrix Regulates Myogenesis. Cells 2019; 8:cells8040332. [PMID: 30970625 PMCID: PMC6523808 DOI: 10.3390/cells8040332] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/06/2019] [Accepted: 04/09/2019] [Indexed: 12/31/2022] Open
Abstract
Dermatopontin (DPT) is an extensively distributed non-collagenous component of the extracellular matrix predominantly found in the dermis of the skin, and consequently expressed in several tissues. In this study, we explored the role of DPT in myogenesis and perceived that it enhances the cell adhesion, reduces the cell proliferation and promotes the myoblast differentiation in C2C12 cells. Our results reveal an inhibitory effect with fibronectin (FN) in myoblast differentiation. We also observed that DPT and fibromodulin (FMOD) regulate positively to each other and promote myogenic differentiation. We further predicted the 3D structure of DPT, which is as yet unknown, and validated it using state-of-the-art in silico tools. Furthermore, we explored the in-silico protein-protein interaction between DPT-FMOD, DPT-FN, and FMOD-FN, and perceived that the interaction between FMOD-FN is more robust than DPT-FMOD and DPT-FN. Taken together, our findings have determined the role of DPT at different stages of the myogenic process.
Collapse
Affiliation(s)
- Taeyeon Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185236, India.
| | - Myung-Gi Seo
- Department of Veterinary Histology, College of Veterinary Medicine, Kyungpook National University, Daegu 702-701, Korea.
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| |
Collapse
|
43
|
Zhang Q, Zhou M, Wu X, Li Z, Liu B, Gao W, Yue J, Liu T. Promoting therapeutic angiogenesis of focal cerebral ischemia using thrombospondin-4 (TSP4) gene-modified bone marrow stromal cells (BMSCs) in a rat model. J Transl Med 2019; 17:111. [PMID: 30947736 PMCID: PMC6449913 DOI: 10.1186/s12967-019-1845-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/11/2019] [Indexed: 01/01/2023] Open
Abstract
Background A stroke caused by angiostenosis always has a poor prognosis. Bone marrow stromal cells (BMSC) are widely applied in vascular regeneration. Recently, thrombospondin-4 (TSP4) was reported to promote the regeneration of blood vessels and enhance the function of endothelial cells in angiogenesis. In this work, we observed the therapeutic effect of TSP4-overexpressing BMSCs on angiogenesis post-stroke. Methods We subcloned the tsp4 gene into a lentivirus expression vector system and harvested the tsp4 lentivirus using 293FT cells. Primary BMSCs were then successfully infected by the tsp4 virus, and overexpression of GFP-fused TSP4 was confirmed by both western blot and immunofluorescence. In vitro, TSP4-overexpressing BMSCs and wild-type BMSCs were co-cultured with human umbilical vein endothelial cells (HUVECs). The expression level of TSP4, vascular endothelial growth factor (VEGF) and transforming growth factor-β (TGF-β) in the supernatant were detected by enzyme-linked immunosorbent assay (ELISA). Wound healing, tube formation and an arterial ring test were performed to estimate the ability of TSP4-overexpressing BMSCs to promote the angiogenesis of endothelial cells. Using a rat permanent middle cerebral artery occlusion (MCAO) model, the effect of TSP4-overexpressing BMSCs on the regeneration of blood vessels was systematically tested by the neurological function score, immunohistochemistry and immunofluorescence staining assays. Results Our results demonstrated that TSP4-overexpressing BMSCs largely increased the expression of VEGF, angiopoietin-1 (Ang-1), matrix metalloprotein 9 (MMP9), matrix metalloprotein 2 (MMP2) and p-Cdc42/Rac1 in endothelial cells. TSP4-BMSC treatment notably up-regulated the TGF-β/Smad2/3 signalling pathway in HUVECs. In vivo, the TSP4-BMSC infusion improved the neurological function score of MCAO rats and expanded the expression of the von Willebrand factor (vWF), Ang-1, MMP2 and MMP9 proteins in cerebral ischemic penumbra. Conclusions Our data illustrate that TSP4-BMSCs can promote the proliferation and migration of endothelial cells and tube formation. We found that TSP4-BMSC infusion can promote the recovery of neural function post-stroke. The tsp4 gene-modified BMSCs provides a better therapeutic effect than that of wild-type BMSCs. Electronic supplementary material The online version of this article (10.1186/s12967-019-1845-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Meiling Zhou
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Xiangfeng Wu
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, People's Republic of China
| | - Zhu Li
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Bing Liu
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Wenbin Gao
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Jin Yue
- The 230th Hospital of the Chinese PLA, Dandong, Liaoning, People's Republic of China.
| | - Tao Liu
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China. .,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China.
| |
Collapse
|
44
|
Zielinski MS, Vardar E, Vythilingam G, Engelhardt EM, Hubbell JA, Frey P, Larsson HM. Quantitative intrinsic auto-cathodoluminescence can resolve spectral signatures of tissue-isolated collagen extracellular matrix. Commun Biol 2019; 2:69. [PMID: 30793047 PMCID: PMC6379429 DOI: 10.1038/s42003-019-0313-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 01/18/2019] [Indexed: 11/18/2022] Open
Abstract
By analyzing isolated collagen gel samples, we demonstrated in situ detection of spectrally deconvoluted auto-cathodoluminescence signatures of specific molecular content with precise spatial localization over a maximum field of view of 300 µm. Correlation of the secondary electron and the hyperspectral images proved ~40 nm resolution in the optical channel, obtained due to a short carrier diffusion length, suppressed by fibril dimensions and poor electrical conductivity specific to their organic composition. By correlating spectrally analyzed auto-cathodoluminescence with mass spectroscopy data, we differentiated spectral signatures of two extracellular matrices, namely human fibrin complex and rat tail collagen isolate, and uncovered differences in protein distributions of isolated extracellular matrix networks of heterogeneous populations. Furthermore, we demonstrated that cathodoluminescence can monitor the progress of a human cell-mediated remodeling process, where human collagenous matrix was deposited within a rat collagenous matrix. The revealed change of the heterogeneous biological composition was confirmed by mass spectroscopy. Zielinski et al. show that quantitative label-free cathodoluminescence-scanning electron microscopy differentiates spectral signatures of two extracellular matrices. This method can monitor the progress of a smooth muscle cell-mediated remodeling process without using antibodies to enhance the optical signal.
Collapse
Affiliation(s)
| | - Elif Vardar
- Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland.,Department of Pediatrics, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, 1011, Switzerland
| | - Ganesh Vythilingam
- Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland.,Department of Surgery, Faculty of Medicine, University Malaya, Kuala Lumpur, 53100, Malaysia
| | - Eva-Maria Engelhardt
- Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Jeffrey A Hubbell
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Peter Frey
- Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Hans M Larsson
- Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland.
| |
Collapse
|
45
|
Mu Y, Zhou DN, Yan NN, Ding JL, Yang J. Upregulation of ADAMTS‑7 and downregulation of COMP are associated with spontaneous abortion. Mol Med Rep 2019; 19:2620-2626. [PMID: 30720083 PMCID: PMC6423623 DOI: 10.3892/mmr.2019.9898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 08/07/2018] [Indexed: 12/27/2022] Open
Abstract
A disintegrin and metalloproteinase with thrombospondin motifs 7 (ADAMTS-7) has been revealed to serve an important role in inflammation-associated diseases. However, the role of ADAMTS-7 in spontaneous abortion (SA) remains unclear. In the present study, human and mouse decidual tissues were used to detect the expression of ADAMTS-7 and cartilage oligomeric matrix protein (COMP) in mice with lipopolysaccharide (LPS)-induced abortion (10 mice/group), and in SA humans and the corresponding control group (21 participants in the SA group and 15 participants in the control group). The results revealed that ADAMTS-7 expression was upregulated and that COMP expression was downregulated in the mouse decidual tissue of the LPS-induced abortion group, when compared with that of the normal control group. The results were further confirmed by western blot analysis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis, which revealed increased ADAMTS-7 and decreased COMP expression at the protein and mRNA levels in mice treated with LPS. Additionally, the expression of ADAMTS-7 was negatively correlated with the expression of COMP in mice, with a correlation coefficient of −0.936 (P<0.001). In addition, the expression of ADAMTS-7 and COMP exhibited was similar in the decidual tissue of SA patients when compared with the levels observed in the tissues of the normal control participants, as demonstrated by increased ADAMTS-7 expression and decreased COMP expression. Western blotting and RT-qPCR analysis revealed that ADAMTS-7 was increased and COMP was decreased in the decidual tissue of SA subjects. The correlation analysis of ADAMTS-7 and COMP in human decidual tissue also revealed a similar result, with a correlation coefficient of −0.836 (P<0.001). The results of the present study demonstrated that ADAMTS-7 was upregulated and COMP was downregulated in the decidual tissues of humans and mice with SA, and a negative correlation was identified between the expression levels of ADAMTS-7 and COMP, thereby providing novel evidence for a better understanding of the pathogenesis of SA, which may lead to improvements in the clinical pregnancy outcomes of these individuals.
Collapse
Affiliation(s)
- Yang Mu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Dan-Ni Zhou
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Na-Na Yan
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jin-Li Ding
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
46
|
Henn D, Abu-Halima M, Wermke D, Falkner F, Thomas B, Köpple C, Ludwig N, Schulte M, Brockmann MA, Kim YJ, Sacks JM, Kneser U, Keller A, Meese E, Schmidt VJ. MicroRNA-regulated pathways of flow-stimulated angiogenesis and vascular remodeling in vivo. J Transl Med 2019; 17:22. [PMID: 30635008 PMCID: PMC6330440 DOI: 10.1186/s12967-019-1767-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 01/02/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Vascular shear stress promotes endothelial cell sprouting in vitro. The impact of hemodynamic forces on microRNA (miRNA) and gene expression within growing vascular networks in vivo, however, remain poorly investigated. Arteriovenous (AV) shunts are an established model for induction of neoangiogenesis in vivo and can serve as a tool for analysis of hemodynamic effects on miRNA and gene expression profiles over time. METHODS AV shunts were microsurgically created in rats and explanted on postoperative days 5, 10 and 15. Neoangiogenesis was confirmed by histologic analysis and micro-computed tomography. MiRNA and gene expression profiles were determined in tissue specimens from AV shunts by microarray analysis and quantitative real-time polymerase chain reaction and compared with sham-operated veins by bioinformatics analysis. Changes in protein expression within AV shunt endothelial cells were determined by immunohistochemistry. RESULTS Samples from AV shunts exhibited a strong overexpression of proangiogenic cytokines, oxygenation-associated genes (HIF1A, HMOX1), and angiopoetic growth factors. Significant inverse correlations of the expressions of miR-223-3p, miR-130b-3p, miR-19b-3p, miR-449a-5p, and miR-511-3p which were up-regulated in AV shunts, and miR-27b-3p, miR-10b-5p, let-7b-5p, and let-7c-5p, which were down-regulated in AV shunts, with their predicted interacting targets C-X-C chemokine receptor 2 (CXCR2), interleukin-1 alpha (IL1A), ephrin receptor kinase 2 (EPHA2), synaptojanin-2 binding protein (SYNJ2BP), forkhead box C1 (FOXC1) were present. CXCL2 and IL1A overexpression in AV shunt endothelium was confirmed at the protein level by immunohistochemistry. CONCLUSIONS Our data indicate that flow-stimulated angiogenesis is determined by an upregulation of cytokines, oxygenation associated genes and miRNA-dependent regulation of FOXC1, EPHA2 and SYNJ2BP.
Collapse
Affiliation(s)
- Dominic Henn
- Department of Hand, Plastic and Reconstructive Surgery, University of Heidelberg, BG Trauma Center Ludwigshafen, Ludwig-Guttmann Str. 13, 67071, Ludwigshafen, Germany
| | - Masood Abu-Halima
- Institute of Human Genetics, Saarland University, Homburg-Saar, Germany
| | - Dominik Wermke
- Institute of Clinical Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Florian Falkner
- Department of Hand, Plastic and Reconstructive Surgery, University of Heidelberg, BG Trauma Center Ludwigshafen, Ludwig-Guttmann Str. 13, 67071, Ludwigshafen, Germany
| | - Benjamin Thomas
- Department of Hand, Plastic and Reconstructive Surgery, University of Heidelberg, BG Trauma Center Ludwigshafen, Ludwig-Guttmann Str. 13, 67071, Ludwigshafen, Germany
| | - Christoph Köpple
- Department of Hand, Plastic and Reconstructive Surgery, University of Heidelberg, BG Trauma Center Ludwigshafen, Ludwig-Guttmann Str. 13, 67071, Ludwigshafen, Germany
| | - Nicole Ludwig
- Institute of Human Genetics, Saarland University, Homburg-Saar, Germany
| | - Matthias Schulte
- Department of Hand, Plastic and Reconstructive Surgery, University of Heidelberg, BG Trauma Center Ludwigshafen, Ludwig-Guttmann Str. 13, 67071, Ludwigshafen, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Center Mainz, Mainz, Germany
| | - Yoo-Jin Kim
- Institute of Pathology, Kaiserslautern, Germany
| | - Justin M Sacks
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ulrich Kneser
- Department of Hand, Plastic and Reconstructive Surgery, University of Heidelberg, BG Trauma Center Ludwigshafen, Ludwig-Guttmann Str. 13, 67071, Ludwigshafen, Germany
| | - Andreas Keller
- Institute of Clinical Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, Homburg-Saar, Germany
| | - Volker J Schmidt
- Department of Hand, Plastic and Reconstructive Surgery, University of Heidelberg, BG Trauma Center Ludwigshafen, Ludwig-Guttmann Str. 13, 67071, Ludwigshafen, Germany.
| |
Collapse
|
47
|
Abstract
Vascular remodeling defines cancer growth and aggressiveness. Although cancer cells produce pro-angiogenic signals, the fate of angiogenesis critically depends on the cancer microenvironment. Composition of the extracellular matrix (ECM) and tumor inflammation determine whether a cancer will remain dormant, will be recognized by the immune system and eliminated, or whether the tumor will develop and lead to the spread and metastasis of cancer cells. Thrombospondins (TSPs), a family of ECM proteins that has long been associated with the regulation of angiogenesis and cancer, regulate multiple physiological processes that determine cancer growth and spreading, from angiogenesis to inflammation, metabolic changes, and properties of ECM. Here, we sought to review publications that describe various functions of TSPs that link these proteins to regulation of cancer growth by modulating multiple physiological and pathological events that prevent or support tumor development. In addition to its direct effects on angiogenesis, TSPs have important roles in regulation of inflammation, immunity, ECM properties and composition, and glucose and insulin metabolism. Furthermore, TSPs have distinct roles as regulators of remodeling in tissues and tumors, such that the pathways activated by a single TSP can interact and influence each other. The complex nature of TSP interactions and functions, including their different cell- and tissue-specific effects, may lead to confusing results and controversial conclusions when taken out of the context of interdisciplinary and holistic approaches. However, studies of TSP functions and roles in different systems of the organism offer an integrative view of tumor remodeling and a potential for finding therapeutic targets that would modulate multiple complementary processes associated with cancer growth.
Collapse
Affiliation(s)
| | - Santoshi Muppala
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, 44195, USA
| | - Jasmine Gajeton
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, 44195, USA
| |
Collapse
|
48
|
Abstract
The immune response to acute muscle damage is important for normal repair. However, in chronic diseases such as many muscular dystrophies, the immune response can amplify pathology and play a major role in determining disease severity. Muscular dystrophies are inheritable diseases that vary tremendously in severity, but share the progressive loss of muscle mass and function that can be debilitating and lethal. Mutations in diverse genes cause muscular dystrophy, including genes that encode proteins that maintain membrane strength, participate in membrane repair, or are components of the extracellular matrix or the nuclear envelope. In this article, we explore the hypothesis that an important feature of many muscular dystrophies is an immune response adapted to acute, infrequent muscle damage that is misapplied in the context of chronic injury. We discuss the involvement of the immune system in the most common muscular dystrophy, Duchenne muscular dystrophy, and show that the immune system influences muscle death and fibrosis as disease progresses. We then present information on immune cell function in other muscular dystrophies and show that for many muscular dystrophies, release of cytosolic proteins into the extracellular space may provide an initial signal, leading to an immune response that is typically dominated by macrophages, neutrophils, helper T-lymphocytes, and cytotoxic T-lymphocytes. Although those features are similar in many muscular dystrophies, each muscular dystrophy shows distinguishing features in the magnitude and type of inflammatory response. These differences indicate that there are disease-specific immunomodulatory molecules that determine response to muscle cell damage caused by diverse genetic mutations. © 2018 American Physiological Society. Compr Physiol 8:1313-1356, 2018.
Collapse
Affiliation(s)
- James G. Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Steven S. Welc
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| |
Collapse
|
49
|
Wang Y, Chen W, Hou B, Gao Y, Li Z, Li X, Zhang C. Altered expression of thrombospondin-1/-2 in the cortex and synaptophysin in the hippocampus after middle cerebral artery occlusion and reperfusion. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3267-3276. [PMID: 31949701 PMCID: PMC6962822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/23/2018] [Indexed: 06/10/2023]
Abstract
Blood supply returned to infracted tissue causes tissue damage. Therefore, ischemia/reperfusion (I/R) injuries are usually accompanied by synapse formation, but the exact cause is still unknown. To address this question, we established a middle cerebral artery occlusion (MCAO) rat model with different reperfusion times, and we examined neurological deficit scores and brain infarct size. Subsequently, thrombospondin (TSP)-1 and TSP-2 expression levels in the cingulated cortex, striate cortex, aud cortex, and piriform cortex at different time points after I/R were examined using immunohistochemistry (IHC). In addition, synaptophysin expression in the hippocampus was examined using IHC. As expected, after ischemia with different reperfusion times, higher neurological deficits scores were observed in MCAO rats compared to sham-operated rats. Brain infarct sizes were increased in different sections, and brain sections exhibited obvious necrosis in the right cerebra. In addition, TSP-1 and TSP-2 expression levels in the cingulated cortex, striate cortex, aud cortex, and piriform cortex significantly increased with increasing reperfusion times. Similarly, synaptophysin expression levels in the hippocampus significantly increased with increasing reperfusion times. Our results indicate that altered TSP-1 and TSP-2 expression in cortical areas may contribute to synapse formation. Our model not only allowed us to observe the time-related expression of TSP-1, TSP-2, and synaptophysin after I/R injury but also provides a potential tool for studying synapse formation.
Collapse
Affiliation(s)
- Yu Wang
- Medical Experiment Center, Shaanxi University of Chinese MedicineXianyang, China
| | - Wei Chen
- Center for Translational Medicine, The First Affiliated Hospital, College of Medicine, Xi’an Jiaotong UniversityXi’an, China
| | - Bin Hou
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center of PLABeijing, China
| | - Yan Gao
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center of PLABeijing, China
| | - Zhihui Li
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center of PLABeijing, China
| | - Xu Li
- Center for Translational Medicine, The First Affiliated Hospital, College of Medicine, Xi’an Jiaotong UniversityXi’an, China
| | - Chenggang Zhang
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center of PLABeijing, China
| |
Collapse
|
50
|
Lin CY, He JY, Zeng CW, Loo MR, Chang WY, Zhang PH, Tsai HJ. microRNA-206 modulates an Rtn4a/Cxcr4a/Thbs3a axis in newly forming somites to maintain and stabilize the somite boundary formation of zebrafish embryos. Open Biol 2018; 7:rsob.170009. [PMID: 28701377 PMCID: PMC5541343 DOI: 10.1098/rsob.170009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
Although microRNA-206 (miR-206) is known to regulate proliferation and differentiation of muscle fibroblasts, the role of miR-206 in early-stage somite development is still unknown. During somitogenesis of zebrafish embryos, reticulon4a (rtn4a) is specifically repressed by miR-206. The somite boundary was defective, and actin filaments were crossing over the boundary in either miR-206-knockdown or rtn4a-overexpressed embryos. In these treated embryos, C-X-C motif chemokine receptor 4a (cxcr4a) was reduced, while thrombospondin 3a (thbs3a) was increased. The defective boundary was phenocopied in either cxcr4a-knockdown or thbs3a-overexpressed embryos. Repression of thbs3a expression by cxcr4a reduced the occurrence of the boundary defect. We demonstrated that cxcr4a is an upstream regulator of thbs3a and that defective boundary cells could not process epithelialization in the absence of intracellular accumulation of the phosphorylated focal adhesion kinase (p-FAK) in boundary cells. Therefore, in the newly forming somites, miR-206-mediated downregulation of rtn4a increases cxcr4a. This activity largely decreases thbs3a expression in the epithelial cells of the somite boundary, which causes epithelialization of boundary cells through mesenchymal-epithelial transition (MET) and eventually leads to somite boundary formation. Collectively, we suggest that miR-206 mediates a novel pathway, the Rtn4a/Cxcr4a/Thbs3a axis, that allows boundary cells to undergo MET and form somite boundaries in the newly forming somites of zebrafish embryos.
Collapse
Affiliation(s)
- Cheng-Yung Lin
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| | - Jun-Yu He
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Chih-Wei Zeng
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Moo-Rumg Loo
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Wen-Yen Chang
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Po-Hsiang Zhang
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| | - Huai-Jen Tsai
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| |
Collapse
|