1
|
Ogata FT, Verma S, Coulson-Thomas VJ, Gesteira TF. TGF-β-Based Therapies for Treating Ocular Surface Disorders. Cells 2024; 13:1105. [PMID: 38994958 PMCID: PMC11240592 DOI: 10.3390/cells13131105] [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: 05/17/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
The cornea is continuously exposed to injuries, ranging from minor scratches to deep traumas. An effective healing mechanism is crucial for the cornea to restore its structure and function following major and minor insults. Transforming Growth Factor-Beta (TGF-β), a versatile signaling molecule that coordinates various cell responses, has a central role in corneal wound healing. Upon corneal injury, TGF-β is rapidly released into the extracellular environment, triggering cell migration and proliferation, the differentiation of keratocytes into myofibroblasts, and the initiation of the repair process. TGF-β-mediated processes are essential for wound closure; however, excessive levels of TGF-β can lead to fibrosis and scarring, causing impaired vision. Three primary isoforms of TGF-β exist-TGF-β1, TGF-β2, and TGF-β3. Although TGF-β isoforms share many structural and functional similarities, they present distinct roles in corneal regeneration, which adds an additional layer of complexity to understand the role of TGF-β in corneal wound healing. Further, aberrant TGF-β activity has been linked to various corneal pathologies, such as scarring and Peter's Anomaly. Thus, understanding the molecular and cellular mechanisms by which TGF-β1-3 regulate corneal wound healing will enable the development of potential therapeutic interventions targeting the key molecule in this process. Herein, we summarize the multifaceted roles of TGF-β in corneal wound healing, dissecting its mechanisms of action and interactions with other molecules, and outline its role in corneal pathogenesis.
Collapse
Affiliation(s)
- Fernando T Ogata
- College of Optometry, University of Houston, 4901 Calhoun Road, Houston, TX 77204, USA
| | - Sudhir Verma
- College of Optometry, University of Houston, 4901 Calhoun Road, Houston, TX 77204, USA
- Deen Dayal Upadhyaya College, University of Delhi, Delhi 110078, India
| | | | - Tarsis F Gesteira
- College of Optometry, University of Houston, 4901 Calhoun Road, Houston, TX 77204, USA
| |
Collapse
|
2
|
Dai G, Sun Y, Wei R, Xi L. Small Leucine-Rich Proteoglycan PODNL1 Identified as a Potential Tumor Matrix-Mediated Biomarker for Prognosis and Immunotherapy in a Pan-Cancer Setting. Curr Issues Mol Biol 2023; 45:6116-6139. [PMID: 37504302 PMCID: PMC10378008 DOI: 10.3390/cimb45070386] [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: 06/27/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023] Open
Abstract
The podocan-like protein 1 (PODNL1), an important member of the small leucine-rich proteoglycans (SLRP) family, is a crucial component of the tumor microenvironment (TME). But its prognostic values and the role in the TME have not been systematically estimated in a pan-cancer setting. Targeting PODNL1, a systematic exploration into the TCGA datasets, reconciling with the analyses of single-cell transcriptomes and immunotherapeutic cohorts in cancers, and validation by tissue microarray-based multiplex immunofluorescence staining was performed. PODNL1 was significantly correlated with the poor prognosis and immunotherapeutic responses in various cancers. In-depth demonstration of molecular mechanisms indicated that PODNL1 expressions were notably positively correlated with cancer-associated fibroblast (CAF) infiltration levels in 33 types of cancers. It also positively correlated with the pan-fibroblast TGF-β response signature score, and the hallmarks including TGF-β, TNF-α, inflammatory response, apical junction, epithelial-mesenchymal transition and hedgehog in pan-cancer. Furthermore, high PODNL1 expressions were positively related with the regulation of tumor-promoting TGF-β signaling through downregulating SMAD2/3:4 heterotrimer regulations transcription and up-regulating the pathway restricted SMAD protein phosphorylation. Single-cell transcriptome analyses and immunofluorescence validations indicated that PODNL1 was predominantly expressed in the cancer cells and CAFs in various cancers. Additionally, the heterogeneity of cancer genotype-phenotype cross-talking was also observed associated with PODNL1. Our systematic study indicates that PODNL1 plays an important role in the complex regulation network of tumor progression, and lays a foundation for further exploration to develop PODNL1 as a valuable matrix-mediated biomarker for cancer immunotherapy and prognosis in a pan-cancer setting.
Collapse
Affiliation(s)
- Geyang Dai
- Cancer Biology Research Center, Key Laboratory of the Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Sun
- Cancer Biology Research Center, Key Laboratory of the Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui Wei
- Cancer Biology Research Center, Key Laboratory of the Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling Xi
- Cancer Biology Research Center, Key Laboratory of the Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
3
|
Zhang JM, Au DT, Sawada H, Franklin MK, Moorleghen JJ, Howatt DA, Wang P, Aicher BO, Hampton B, Migliorini M, Ni F, Mullick AE, Wani MM, Ucuzian AA, Lu HS, Muratoglu SC, Daugherty A, Strickland DK. LRP1 protects against excessive superior mesenteric artery remodeling by modulating angiotensin II-mediated signaling. JCI Insight 2023; 8:e164751. [PMID: 36472907 PMCID: PMC9977308 DOI: 10.1172/jci.insight.164751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (vSMCs) exert a critical role in sensing and maintaining vascular integrity. These cells abundantly express the low-density lipoprotein receptor-related protein 1 (LRP1), a large endocytic signaling receptor that recognizes numerous ligands, including apolipoprotein E-rich lipoproteins, proteases, and protease-inhibitor complexes. We observed the spontaneous formation of aneurysms in the superior mesenteric artery (SMA) of both male and female mice in which LRP1 was genetically deleted in vSMCs (smLRP1-/- mice). Quantitative proteomics revealed elevated abundance of several proteins in smLRP1-/- mice that are known to be induced by angiotensin II-mediated (AngII-mediated) signaling, suggesting that this pathway was dysregulated. Administration of losartan, an AngII type I receptor antagonist, or an angiotensinogen antisense oligonucleotide to reduce plasma angiotensinogen concentrations restored the normal SMA phenotype in smLRP1-/- mice and prevented aneurysm formation. Additionally, using a vascular injury model, we noted excessive vascular remodeling and neointima formation in smLRP1-/- mice that was restored by losartan administration. Together, these findings reveal that LRP1 regulates vascular integrity and remodeling of the SMA by attenuating excessive AngII-mediated signaling.
Collapse
Affiliation(s)
- Jackie M Zhang
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dianaly T Au
- Center for Vascular and Inflammatory Diseases and
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center and
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | | | | | | | - Pengjun Wang
- Saha Cardiovascular Research Center and Saha Aortic Center and
| | - Brittany O Aicher
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | - Fenge Ni
- Center for Vascular and Inflammatory Diseases and
| | | | | | - Areck A Ucuzian
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Vascular Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| | - Hong S Lu
- Saha Cardiovascular Research Center and Saha Aortic Center and
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | | | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center and
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Matthews EZ, Lanham S, White K, Kyriazi ME, Alexaki K, El-Sagheer AH, Brown T, Kanaras AG, J West J, MacArthur BD, Stumpf PS, Oreffo ROC. Single-cell RNA-sequence analysis of human bone marrow reveals new targets for isolation of skeletal stem cells using spherical nucleic acids. J Tissue Eng 2023; 14:20417314231169375. [PMID: 37216034 PMCID: PMC10192814 DOI: 10.1177/20417314231169375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/24/2023] [Indexed: 05/24/2023] Open
Abstract
There is a wealth of data indicating human bone marrow contains skeletal stem cells (SSC) with the capacity for osteogenic, chondrogenic and adipogenic differentiation. However, current methods to isolate SSCs are restricted by the lack of a defined marker, limiting understanding of SSC fate, immunophenotype, function and clinical application. The current study applied single-cell RNA-sequencing to profile human adult bone marrow populations from 11 donors and identified novel targets for SSC enrichment. Spherical nucleic acids were used to detect these mRNA targets in SSCs. This methodology was able to rapidly isolate potential SSCs found at a frequency of <1 in 1,000,000 in human bone marrow, with the capacity for tri-lineage differentiation in vitro and ectopic bone formation in vivo. The current studies detail the development of a platform to advance SSC enrichment from human bone marrow, offering an invaluable resource for further SSC characterisation, with significant therapeutic impact therein.
Collapse
Affiliation(s)
- Elloise Z Matthews
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
| | - Stuart Lanham
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Cancer Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
| | - Kate White
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
| | - Maria-Eleni Kyriazi
- College of Engineering and Technology,
American University of the Middle East, Kuwait
| | - Konstantina Alexaki
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Afaf H El-Sagheer
- Department of Chemistry, Chemistry
Research Laboratory, University of Oxford, Oxford, UK
- Chemistry Branch, Department of Science
and Mathematics, Faculty of Petroleum and Mining Engineering, Suez University, Suez,
Egypt
| | - Tom Brown
- Department of Chemistry, Chemistry
Research Laboratory, University of Oxford, Oxford, UK
| | - Antonios G Kanaras
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
| | - Jonathan J West
- Cancer Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Ben D MacArthur
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
- Mathematical Sciences, University of
Southampton, Southampton, UK
| | - Patrick S Stumpf
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Joint Research Center for Computational
Biomedicine, RWTH Aachen University, Aachen, Germany
| | - Richard OC Oreffo
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
- College of Biomedical Engineering,
China Medical University, Taichung, Taiwan
| |
Collapse
|
5
|
Urbanczyk M, Jeyagaran A, Zbinden A, Lu CE, Marzi J, Kuhlburger L, Nahnsen S, Layland SL, Duffy G, Schenke-Layland K. Decorin improves human pancreatic β-cell function and regulates ECM expression in vitro. Matrix Biol 2023; 115:160-183. [PMID: 36592738 DOI: 10.1016/j.matbio.2022.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
Transplantation of islets of Langerhans is a promising alternative treatment strategy in severe cases of type 1 diabetes mellitus; however, the success rate is limited by the survival rate of the cells post-transplantation. Restoration of the native pancreatic niche during transplantation potentially can help to improve cell viability and function. Here, we assessed for the first time the regulatory role of the small leucine-rich proteoglycan decorin (DCN) in insulin secretion in human β-cells, and its impact on pancreatic extracellular matrix (ECM) protein expression in vitro. In depth analyses utilizing next-generation sequencing as well as Raman microspectroscopy and Raman imaging identified pathways related to glucose metabolism to be upregulated in DCN-treated cells, including oxidative phosphorylation within the mitochondria as well as proteins and lipids of the endoplasmic reticulum. We further showed the effectiveness of DCN in a transplantation setting by treating collagen type 1-encapsulated β-cell-containing pseudo-islets with DCN. Taken together, in this study, we demonstrate the potential of DCN to improve the function of insulin-secreting β-cells while reducing the expression of ECM proteins affiliated with fibrotic capsule formation, making DCN a highly promising therapeutic agent for islet transplantation.
Collapse
Affiliation(s)
- Max Urbanczyk
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Silcherstr. 7/1, Tübingen 72076, Germany
| | - Abiramy Jeyagaran
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Silcherstr. 7/1, Tübingen 72076, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Aline Zbinden
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Silcherstr. 7/1, Tübingen 72076, Germany; Department of Immunology, Leiden University Medical Center Leiden, ZA 2333, the Netherlands
| | - Chuan-En Lu
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Silcherstr. 7/1, Tübingen 72076, Germany
| | - Julia Marzi
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Silcherstr. 7/1, Tübingen 72076, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Tübingen, Germany
| | - Laurence Kuhlburger
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany; Biomedical Data Science, Department of Computer Science, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany; Biomedical Data Science, Department of Computer Science, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Shannon L Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Silcherstr. 7/1, Tübingen 72076, Germany; Department of Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Garry Duffy
- Discipline of Anatomy and the Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Advanced Materials for Biomedical Engineering (AMBER), Trinity College Dublin & National University of Ireland Galway, Galway, Ireland
| | - Katja Schenke-Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Silcherstr. 7/1, Tübingen 72076, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Tübingen, Germany.
| |
Collapse
|
6
|
Kubo E, Shibata S, Shibata T, Sasaki H, Singh DP. Role of Decorin in the Lens and Ocular Diseases. Cells 2022; 12:cells12010074. [PMID: 36611867 PMCID: PMC9818407 DOI: 10.3390/cells12010074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Decorin is an archetypal member of the small leucine-rich proteoglycan gene family and is involved in various biological functions and many signaling networks, interacting with extra-cellular matrix (ECM) components, growth factors, and receptor tyrosine kinases. Decorin also modulates the growth factors, cell proliferation, migration, and angiogenesis. It has been reported to be involved in many ischemic and fibrotic eye diseases, such as congenital stromal dystrophy of the cornea, anterior subcapsular fibrosis of the lens, proliferative vitreoretinopathy, et al. Furthermore, recent evidence supports its role in secondary posterior capsule opacification (PCO) after cataract surgery. The expression of decorin mRNA in lens epithelial cells in vitro was found to decrease upon transforming growth factor (TGF)-β-2 addition and increase upon fibroblast growth factor (FGF)-2 addition. Wound healing of the injured lens in mice transgenic for lens-specific human decorin was promoted by inhibiting myofibroblastic changes. Decorin may be associated with epithelial-mesenchymal transition and PCO development in the lens. Gene therapy and decorin administration have the potential to serve as excellent therapeutic approaches for modifying impaired wound healing, PCO, and other eye diseases related to fibrosis and angiogenesis. In this review, we present findings regarding the roles of decorin in the lens and ocular diseases.
Collapse
Affiliation(s)
- Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 920-0293, Ishikawa, Japan
- Correspondence: ; Tel.: +81-76-286-2211 (ext. 3412); Fax: +81-76-286-1010
| | - Shinsuke Shibata
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 920-0293, Ishikawa, Japan
| | - Teppei Shibata
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 920-0293, Ishikawa, Japan
| | - Hiroshi Sasaki
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 920-0293, Ishikawa, Japan
| | - Dhirendra P. Singh
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
7
|
Singh K, Sachan N, Ene T, Dabovic B, Rifkin D. Latent Transforming Growth Factor β Binding Protein 3 Controls Adipogenesis. Matrix Biol 2022; 112:155-170. [PMID: 35933071 DOI: 10.1016/j.matbio.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022]
Abstract
Transforming growth factor-beta (TGFβ) is released from cells as part of a trimeric latent complex consisting of TGFβ, the TGFβ propeptides, and either a latent TGFβ binding protein (LTBP) or glycoprotein-A repetitions predominant (GARP) protein. LTBP1 and 3 modulate latent TGFβ function with respect to secretion, matrix localization, and activation and, therefore, are vital for the proper function of the cytokine in a number of tissues. TGFβ modulates stem cell differentiation into adipocytes (adipogenesis), but the potential role of LTBPs in this process has not been studied. We observed that 72 h post adipogenesis initiation Ltbp1, 2, and 4 expression levels decrease by 74-84%, whereas Ltbp3 expression levels remain constant during adipogenesis. We found that LTBP3 silencing in C3H/10T1/2 cells reduced adipogenesis, as measured by the percentage of cells with lipid vesicles and the expression of the transcription factor peroxisome proliferator-activated receptor gamma (PPARγ). Lentiviral mediated expression of an Ltbp3 mRNA resistant to siRNA targeting rescued the phenotype, validating siRNA specificity. Knockdown (KD) of Ltbp3 expression in 3T3-L1, M2, and primary bone marrow stromal cells (BMSC) indicated a similar requirement for Ltbp3. Epididymal and inguinal white adipose tissue fat pad weights of Ltbp3-/- mice were reduced by 62% and 57%, respectively, compared to wild-type mice. Inhibition of adipogenic differentiation upon LTBP3 loss is mediated by TGFβ, as TGFβ neutralizing antibody and TGFβ receptor I kinase blockade rescue the LTBP3 KD phenotype. These results indicate that LTBP3 has a TGFβ-dependent function in adipogenesis both in vitro and possibly in vivo.
Collapse
Affiliation(s)
- Karan Singh
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nalani Sachan
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Taylor Ene
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Branka Dabovic
- Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Daniel Rifkin
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA; Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Mogensen EH, Poulsen ET, Thøgersen IB, Yamamoto K, Brüel A, Enghild JJ. The low-density lipoprotein receptor-related protein 1 (LRP1) interactome in the human cornea. Exp Eye Res 2022; 219:109081. [PMID: 35461874 DOI: 10.1016/j.exer.2022.109081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/27/2022] [Accepted: 04/17/2022] [Indexed: 12/25/2022]
Abstract
The human cornea is responsible for approximately 70% of the eye's optical power and, together with the lens, constitutes the only transparent tissue in the human body. Low-density lipoprotein receptor-related protein 1 (LRP1), a large, multitalented endocytic receptor, is expressed throughout the human cornea, yet its role in the cornea remains unknown. More than 30 years ago, LRP1 was purified by exploiting its affinity for the activated form of the protease inhibitor alpha-2-macroblulin (A2M), and the original purification protocol is generally referred to in studies involving full-length LRP1. Here, we provide a novel and simplified LRP1 purification protocol based on LRP1's affinity for receptor-related protein (RAP) that produces significantly higher yields of authentic LRP1. Purified LRP1 was used to map its unknown interactome in the human cornea. Corneal proteins extracted under physiologically relevant conditions were subjected to LRP1 affinity pull-down, and LRP1 ligand candidates were identified by LC-MS/MS. A total of 28 LRP1 ligand candidates were found, including 22 novel ligands. The LRP1 corneal interactome suggests a novel role for LRP1 as a regulator of the corneal immune response, structure, and ultimately corneal transparency.
Collapse
Affiliation(s)
- Emilie Hage Mogensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Ida B Thøgersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
9
|
Xie C, Mondal DK, Ulas M, Neill T, Iozzo RV. Oncosuppressive roles of decorin through regulation of multiple receptors and diverse signaling pathways. Am J Physiol Cell Physiol 2022; 322:C554-C566. [PMID: 35171698 PMCID: PMC8917911 DOI: 10.1152/ajpcell.00016.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Decorin is a stromal-derived prototype member of the small leucine-rich proteoglycan gene family. In addition to its functions as a regulator of collagen fibrillogenesis and TGF-β activity soluble decorin acts as a pan-receptor tyrosine kinase (RTK) inhibitor. Decorin binds to various RTKs including EGFR HER2 HGFR/Met VEGFR2 TLR and IGFR. Although the molecular mechanism for the action of decorin on these receptors is not entirely elucidated overall decorin evokes transient activation of these receptors with suppression of downstream signaling cascades culminating in growth inhibition followed by their physical downregulation via caveosomal internalization and degradation. In the case of Met decorin leads to decreased β-catenin signaling pathway and growth suppression. As most of these RTKs are responsible for providing a growth advantage to cancer cells the result of decorin treatment is oncosuppression. Another decorin-driven mechanism to restrict cancer growth and dissemination is by impeding angiogenesis via vascular endothelial growth factor receptor 2 (VEGFR2) and the concurrent activation of protracted endothelial cell autophagy. In this review we will dissect the multiple roles of decorin in cancer biology and its potential use as a next-generation protein-based adjuvant therapy to combat cancer.
Collapse
Affiliation(s)
- Christopher Xie
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Dipon K. Mondal
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mikdat Ulas
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Nitahara-Kasahara Y, Posadas-Herrera G, Mizumoto S, Nakamura-Takahashi A, Inoue YU, Inoue T, Nomura Y, Takeda S, Yamada S, Kosho T, Okada T. Myopathy Associated With Dermatan Sulfate-Deficient Decorin and Myostatin in Musculocontractural Ehlers-Danlos Syndrome: A Mouse Model Investigation. Front Cell Dev Biol 2021; 9:695021. [PMID: 34708033 PMCID: PMC8542786 DOI: 10.3389/fcell.2021.695021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Carbohydrate sulfotransferase 14 (CHST14) encodes dermatan 4-O-sulfotransferase 1, a critical enzyme for dermatan sulfate (DS) biosynthesis. Musculocontractural Ehlers-Danlos syndrome (mcEDS) is associated with biallelic pathogenic variants of CHST14 and is characterized by malformations and manifestations related to progressive connective tissue fragility. We identified myopathy phenotypes in Chst14-deficient mice using an mcEDS model. Decorin is a proteoglycan harboring a single glycosaminoglycan chain containing mainly DS, which are replaced with chondroitin sulfate (CS) in mcEDS patients with CHST14 deficiency. We studied the function of decorin in the skeletal muscle of Chst14-deficient mice because decorin is important for collagen-fibril assembly and has a myokine role in promoting muscle growth. Although decorin was present in the muscle perimysium of wild-type (Chst14+/+ ) mice, decorin was distributed in the muscle perimysium as well as in the endomysium of Chst14-/- mice. Chst14-/- mice had small muscle fibers within the spread interstitium; however, histopathological findings indicated milder myopathy in Chst14-/- mice. Myostatin, a negative regulator of protein synthesis in the muscle, was upregulated in Chst14-/- mice. In the muscle of Chst14-/- mice, decorin was downregulated compared to that in Chst14+/+ mice. Chst14-/- mice showed altered cytokine/chemokine balance and increased fibrosis, suggesting low myogenic activity in DS-deficient muscle. Therefore, DS deficiency in mcEDS causes pathological localization and functional abnormalities of decorin, which causes disturbances in skeletal muscle myogenesis.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Guillermo Posadas-Herrera
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | | | - Yukiko U. Inoue
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan
| | - Takayoshi Inoue
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan
| | - Yoshihiro Nomura
- Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Shin’ichi Takeda
- National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Tomoki Kosho
- Department of Medical Genetics, Shinshu University School of Medicine, Matsumoto, Japan
- Center for Medical Genetics, Shinshu University Hospital, Matsumoto, Japan
- Division of Clinical Sequencing, Shinshu University School of Medicine, Matsumoto, Japan
- Research Center for Supports to Advanced Science, Shinshu University, Matsumoto, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Sepúlveda V, Maurelia F, González M, Aguayo J, Caprile T. SCO-spondin, a giant matricellular protein that regulates cerebrospinal fluid activity. Fluids Barriers CNS 2021; 18:45. [PMID: 34600566 PMCID: PMC8487547 DOI: 10.1186/s12987-021-00277-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/11/2021] [Indexed: 12/28/2022] Open
Abstract
Cerebrospinal fluid is a clear fluid that occupies the ventricular and subarachnoid spaces within and around the brain and spinal cord. Cerebrospinal fluid is a dynamic signaling milieu that transports nutrients, waste materials and neuroactive substances that are crucial for the development, homeostasis and functionality of the central nervous system. The mechanisms that enable cerebrospinal fluid to simultaneously exert these homeostatic/dynamic functions are not fully understood. SCO-spondin is a large glycoprotein secreted since the early stages of development into the cerebrospinal fluid. Its domain architecture resembles a combination of a matricellular protein and the ligand-binding region of LDL receptor family. The matricellular proteins are a group of extracellular proteins with the capacity to interact with different molecules, such as growth factors, cytokines and cellular receptors; enabling the integration of information to modulate various physiological and pathological processes. In the same way, the LDL receptor family interacts with many ligands, including β-amyloid peptide and different growth factors. The domains similarity suggests that SCO-spondin is a matricellular protein enabled to bind, modulate, and transport different cerebrospinal fluid molecules. SCO-spondin can be found soluble or polymerized into a dynamic threadlike structure called the Reissner fiber, which extends from the diencephalon to the caudal tip of the spinal cord. Reissner fiber continuously moves caudally as new SCO-spondin molecules are added at the cephalic end and are disaggregated at the caudal end. This movement, like a conveyor belt, allows the transport of the bound molecules, thereby increasing their lifespan and action radius. The binding of SCO-spondin to some relevant molecules has already been reported; however, in this review we suggest more than 30 possible binding partners, including peptide β-amyloid and several growth factors. This new perspective characterizes SCO-spondin as a regulator of cerebrospinal fluid activity, explaining its high evolutionary conservation, its apparent multifunctionality, and the lethality or severe malformations, such as hydrocephalus and curved body axis, of knockout embryos. Understanding the regulation and identifying binding partners of SCO-spondin are crucial for better comprehension of cerebrospinal fluid physiology.
Collapse
Affiliation(s)
- Vania Sepúlveda
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Felipe Maurelia
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Maryori González
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jaime Aguayo
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Teresa Caprile
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
12
|
Joyce K, Fabra GT, Bozkurt Y, Pandit A. Bioactive potential of natural biomaterials: identification, retention and assessment of biological properties. Signal Transduct Target Ther 2021; 6:122. [PMID: 33737507 PMCID: PMC7973744 DOI: 10.1038/s41392-021-00512-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/29/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Biomaterials have had an increasingly important role in recent decades, in biomedical device design and the development of tissue engineering solutions for cell delivery, drug delivery, device integration, tissue replacement, and more. There is an increasing trend in tissue engineering to use natural substrates, such as macromolecules native to plants and animals to improve the biocompatibility and biodegradability of delivered materials. At the same time, these materials have favourable mechanical properties and often considered to be biologically inert. More importantly, these macromolecules possess innate functions and properties due to their unique chemical composition and structure, which increase their bioactivity and therapeutic potential in a wide range of applications. While much focus has been on integrating these materials into these devices via a spectrum of cross-linking mechanisms, little attention is drawn to residual bioactivity that is often hampered during isolation, purification, and production processes. Herein, we discuss methods of initial material characterisation to determine innate bioactivity, means of material processing including cross-linking, decellularisation, and purification techniques and finally, a biological assessment of retained bioactivity of a final product. This review aims to address considerations for biomaterials design from natural polymers, through the optimisation and preservation of bioactive components that maximise the inherent bioactive potency of the substrate to promote tissue regeneration.
Collapse
Affiliation(s)
- Kieran Joyce
- School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Georgina Targa Fabra
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Yagmur Bozkurt
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland.
| |
Collapse
|
13
|
Molecular Mechanisms of Renal Progenitor Regulation: How Many Pieces in the Puzzle? Cells 2021; 10:cells10010059. [PMID: 33401654 PMCID: PMC7823786 DOI: 10.3390/cells10010059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/26/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Kidneys of mice, rats and humans possess progenitors that maintain daily homeostasis and take part in endogenous regenerative processes following injury, owing to their capacity to proliferate and differentiate. In the glomerular and tubular compartments of the nephron, consistent studies demonstrated that well-characterized, distinct populations of progenitor cells, localized in the parietal epithelium of Bowman capsule and scattered in the proximal and distal tubules, could generate segment-specific cells in physiological conditions and following tissue injury. However, defective or abnormal regenerative responses of these progenitors can contribute to pathologic conditions. The molecular characteristics of renal progenitors have been extensively studied, revealing that numerous classical and evolutionarily conserved pathways, such as Notch or Wnt/β-catenin, play a major role in cell regulation. Others, such as retinoic acid, renin-angiotensin-aldosterone system, TLR2 (Toll-like receptor 2) and leptin, are also important in this process. In this review, we summarize the plethora of molecular mechanisms directing renal progenitor responses during homeostasis and following kidney injury. Finally, we will explore how single-cell RNA sequencing could bring the characterization of renal progenitors to the next level, while knowing their molecular signature is gaining relevance in the clinic.
Collapse
|
14
|
Inubushi T, Kosai A, Yanagisawa S, Chanbora C, Miyauchi M, Yamasaki S, Sugiyama E, Ishikado A, Makino T, Takata T. Bovine lactoferrin enhances osteogenesis through Smad2/3 and p38 MAPK activation. J Oral Biosci 2020; 62:147-154. [DOI: 10.1016/j.job.2020.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 10/24/2022]
|
15
|
Ravalli F, Kossar AP, Takayama H, Grau JB, Ferrari G. Aortic Valve Regurgitation: Pathophysiology and Implications for Surgical Intervention in the Era of TAVR. STRUCTURAL HEART : THE JOURNAL OF THE HEART TEAM 2020; 4:87-98. [PMID: 32529168 PMCID: PMC7288848 DOI: 10.1080/24748706.2020.1719446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 10/25/2022]
Abstract
Aortic insufficiency (AI) or regurgitation is caused by the malcoaptation of the aortic valve (AV) cusps due to intrinsic abnormalities of the valve itself, a dilatation or geometric distortion of the aortic root, or by some combination thereof. In recent years, there has been an increase in the number of studies suggesting that AI is an active disease process caused by a combination of factors including but not limited to alteration of specific molecular pathways, genetic predisposition, and changes in the mechanotransductive properties of the AV apparatus. As the surgical management of AV disease continues to evolve, increasingly sophisticated surgical and percutaneous techniques for AV repair and replacement, including transcatheter aortic valve replacement (TAVR), have become more commonplace and will likely continue to expand as new devices are introduced. However, these techniques necessitate frequent reappraisal of the biological and mechanobiological mechanisms underlying AV regurgitation to better understand the risk factors for AI development and recurrence following surgical intervention as well as expand our limited knowledge on patient selection for such procedures. The aim of this review is to describe some of the putative mechanisms implicated in the development of AI, dissect some of the cross-talk among known and possible signaling pathways leading to valve remodeling, identify association between these pathways and pharmacological approaches, and discuss the implications for surgical and percutaneous approaches to AV repair in replacement in the TAVR era.
Collapse
|
16
|
Au DT, Arai AL, Fondrie WE, Muratoglu SC, Strickland DK. Role of the LDL Receptor-Related Protein 1 in Regulating Protease Activity and Signaling Pathways in the Vasculature. Curr Drug Targets 2019; 19:1276-1288. [PMID: 29749311 DOI: 10.2174/1389450119666180511162048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 12/22/2022]
Abstract
Aortic aneurysms represent a significant clinical problem as they largely go undetected until a rupture occurs. Currently, an understanding of mechanisms leading to aneurysm formation is limited. Numerous studies clearly indicate that vascular smooth muscle cells play a major role in the development and response of the vasculature to hemodynamic changes and defects in these responses can lead to aneurysm formation. The LDL receptor-related protein 1 (LRP1) is major smooth muscle cell receptor that has the capacity to mediate the endocytosis of numerous ligands and to initiate and regulate signaling pathways. Genetic evidence in humans and mouse models reveal a critical role for LRP1 in maintaining the integrity of the vasculature. Understanding the mechanisms by which this is accomplished represents an important area of research, and likely involves LRP1's ability to regulate levels of proteases known to degrade the extracellular matrix as well as its ability to modulate signaling events.
Collapse
Affiliation(s)
- Dianaly T Au
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - Allison L Arai
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - William E Fondrie
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - Selen C Muratoglu
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States
| |
Collapse
|
17
|
Rebolledo DL, González D, Faundez-Contreras J, Contreras O, Vio CP, Murphy-Ullrich JE, Lipson KE, Brandan E. Denervation-induced skeletal muscle fibrosis is mediated by CTGF/CCN2 independently of TGF-β. Matrix Biol 2019; 82:20-37. [DOI: 10.1016/j.matbio.2019.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 02/06/2023]
|
18
|
Zhang Y, Ye T, Gong S, Hong Z, Zhou X, Liu H, Qu H, Qian J. RNA-sequencing based bone marrow cell transcriptome analysis reveals the potential mechanisms of E'jiao against blood-deficiency in mice. Biomed Pharmacother 2019; 118:109291. [PMID: 31401395 DOI: 10.1016/j.biopha.2019.109291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/20/2019] [Accepted: 07/31/2019] [Indexed: 02/05/2023] Open
Abstract
As a health-care food and traditional Chinese medicine, E'jiao, from the skin of Equus animus L, has been used to nourish blood in China for more than 2000 years. In modern medicine, there are also evidences indicate it has a beneficial effect on chemotherapy-caused blood deficiency. However, its mechanism of action for blood invigoration remains unclear. In the present study, we investigated the hematopoietic effect of E'jiao in 5-Fluorouracil-treated mice. In addition to the counting of bone marrow nucleated cells (BMNCs), flow cytometry was used to detect the population of hematopoietic stem cells (HSCs), and colony-forming unit (CFU) was used to assay the differentiation ability of hematopoietic progenitor cells (HPCs). Gene expression profiles of bone marrow cells were obtained from RNA sequencing (RNA-seq) and differentially expressed genes (DEGs) were analyzed with an emphasis on hematopoiesis-related pathways. The results show that E'jiao promotes the proliferation of both BMNCs and HSCs, as well as the differentiation of HPCs. By providing a hematopoiesis-related molecular regulatory network of E'jiao, we point out that the mechanism of E'jiao is associated with pathways including ECM-receptor interaction, Wnt signaling pathway, PI3K-Akt signaling pathway, TGF-beta signaling pathway, Hematopoietic cell lineage and Osteoclast differentiation, in which Ibsp, Col1a1, Col1a2, Notum, Sost, Dkk1, Irx5, Irx3 and Dcn are the key regulatory molecules. These findings provide valuable molecular basis for the mechanism of action of E'jiao.
Collapse
Affiliation(s)
- Yan Zhang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co., Ltd., Liaocheng, China
| | - Tingting Ye
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shuqing Gong
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhuping Hong
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiangshan Zhou
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co., Ltd., Liaocheng, China
| | - Haibin Liu
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co., Ltd., Liaocheng, China.
| | - Haibin Qu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Jing Qian
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
19
|
Potere N, Del Buono MG, Mauro AG, Abbate A, Toldo S. Low Density Lipoprotein Receptor-Related Protein-1 in Cardiac Inflammation and Infarct Healing. Front Cardiovasc Med 2019; 6:51. [PMID: 31080804 PMCID: PMC6497734 DOI: 10.3389/fcvm.2019.00051] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/09/2019] [Indexed: 01/07/2023] Open
Abstract
Acute myocardial infarction (AMI) leads to myocardial cell death and ensuing sterile inflammatory response, which represents an attempt to clear cellular debris and promote cardiac repair. However, an overwhelming, unopposed or unresolved inflammatory response following AMI leads to further injury, worse remodeling and heart failure (HF). Additional therapies are therefore warranted to blunt the inflammatory response associated with ischemia and reperfusion and prevent long-term adverse events. Low-density lipoprotein receptor-related protein 1 (LRP1) is a ubiquitous endocytic cell surface receptor with the ability to recognize a wide range of structurally and functionally diverse ligands. LRP1 transduces multiple intracellular signal pathways regulating the inflammatory reaction, tissue remodeling and cell survival after organ injury. In preclinical studies, activation of LRP1-mediated signaling in the heart with non-selective and selective LRP1 agonists is linked with a powerful cardioprotective effect, reducing infarct size and cardiac dysfunction after AMI. The data from early phase clinical studies with plasma-derived α1-antitrypsin (AAT), an endogenous LRP1 agonist, and SP16 peptide, a synthetic LRP1 agonist, support the translational value of LRP1 as a novel therapeutic target in AMI. In this review, we will summarize the cellular and molecular bases of LRP1 functions in modulating the inflammatory reaction and the reparative process after injury in various peripheral tissues, and discuss recent evidences implicating LRP1 in myocardial inflammation and infarct healing.
Collapse
Affiliation(s)
- Nicola Potere
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Adolfo Gabriele Mauro
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
20
|
Potere N, Del Buono MG, Niccoli G, Crea F, Toldo S, Abbate A. Developing LRP1 Agonists into a Therapeutic Strategy in Acute Myocardial Infarction. Int J Mol Sci 2019; 20:E544. [PMID: 30696029 PMCID: PMC6387161 DOI: 10.3390/ijms20030544] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 01/25/2019] [Indexed: 12/16/2022] Open
Abstract
Cardioprotection refers to a strategy aimed at enhancing survival pathways in the injured yet salvageable myocardium following ischemia-reperfusion. Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor that can be targeted following reperfusion, to induce a cardioprotective signaling through the activation of the reperfusion injury salvage kinase (RISK) pathway. The data from preclinical studies with non-selective and selective LRP1 agonists are promising, showing a large therapeutic window for intervention to reduce infarct size after ischemia-reperfusion. A pilot clinical trial with plasma derived α1-antitrypsin (AAT), a naturally occurring LRP1 agonist, supports the translational value of LRP1 as a novel therapeutic target for cardioprotection. A phase I study with a selective LRP1 agonist has been completed showing no toxicity. These findings may open the way to early phase clinical studies with pharmacologic LRP1 activation in patients with acute myocardial infarction (AMI).
Collapse
Affiliation(s)
- Nicola Potere
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
- Unit of Cardiovascular Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy.
| | - Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Giampaolo Niccoli
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
21
|
Vu TT, Marquez J, Le LT, Nguyen ATT, Kim HK, Han J. The role of decorin in cardiovascular diseases: more than just a decoration. Free Radic Res 2018; 52:1210-1219. [PMID: 30468093 DOI: 10.1080/10715762.2018.1516285] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Decorin (DCN) is a proteoglycan constituent of the extracellular matrix (ECM) possessing powerful antifibrotic, anti-inflammation, antioxidant, and antiangiogenic properties. By attaching to receptors in the cell surface or to several ECM molecules, it regulates plenty of cellular functions, consequently influencing cell differentiation, proliferation, and apoptosis. These processes are dependent on cell types, biological contexts, and interfere with pathological processes such as cardiovascular diseases. In this review, we briefly discuss the potential of DCN targeting in addressing cardiovascular diseases (CVD). We dive into its interactome and discuss how its interaction with the proteins can affect disease progression, and how DCN can be a possible target for CVD therapeutics.
Collapse
Affiliation(s)
- Thu Thi Vu
- a Faculty of Biology, National Key Laboratory of Enzyme and Protein Technology , VNU University of Science , Hanoi , Vietnam
| | - Jubert Marquez
- b National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea.,c National Research Laboratory for Mitochondrial Signaling, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea
| | - Long Thanh Le
- b National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea.,c National Research Laboratory for Mitochondrial Signaling, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea
| | - Anh Thi Tuyet Nguyen
- b National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea.,c National Research Laboratory for Mitochondrial Signaling, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea
| | - Hyoung Kyu Kim
- b National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea.,c National Research Laboratory for Mitochondrial Signaling, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea.,d Department of Integrated Biomedical Science , College of Medicine, Inje University , Busan , Korea
| | - Jin Han
- b National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea.,c National Research Laboratory for Mitochondrial Signaling, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center , College of Medicine, Inje University , Busan , Korea
| |
Collapse
|
22
|
Russo TA, Stoll D, Nader HB, Dreyfuss JL. Mechanical stretch implications for vascular endothelial cells: Altered extracellular matrix synthesis and remodeling in pathological conditions. Life Sci 2018; 213:214-225. [PMID: 30343127 DOI: 10.1016/j.lfs.2018.10.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/09/2018] [Accepted: 10/17/2018] [Indexed: 10/28/2022]
Abstract
AIMS Cardiovascular diseases such as hypertension, thrombosis and atherosclerosis are responses to mechanical forces applied to the endothelium. Endothelial cells respond to hemodynamic mechanical forces such as cellular mechanical stretching. We investigated the expression of glycosaminoglycans, proteoglycans and other extracellular matrix molecules in endothelial cells subjected to various mechanical stimuli. MAIN METHODS Endothelial cells were subjected to mechanical stretch in a vacuum system FlexCell™ to 5% (physiological condition) and 15% (pathological condition), for 4 h or 24 h. Culture plates not subjected to strain were used as controls. Subsequently, ECs were subjected to immunofluorescence, real-time PCR, PCR array, glycosaminoglycans biosynthesis using metabolic radiolabeling with 35S-sulfate and cell behavior assays (adhesion, migration and capillary tube formation). KEY FINDINGS Mechanical stretch induced changes in endothelial cell morphology. Pathological consequences of mechanical stretch included inhibited migration in 2-fold and capillary-like tube formation in 2-fold, when compared to physiological condition after 4 h of ECs exposure; it also reduced total sulfated glycosaminoglycans synthesis thereabout 1.5-fold. Pathological mechanical stretch conditions induced higher expression after 24 h of ECs exposure to mechanical stretch of syndecan-4 (3.5-fold), perlecan (9.1-fold), decorin (5.7-fold), adhesive proteins as fibronectin (5.6-fold) and collagen III α1 (2.2-fold) and growth factors, including VEGF-A (7.3-fold) and TGFβ-1 (14.6-fold) and TGFβ-3 (4.3-fold). SIGNIFICANCE Exposure of endothelial cells to mechanical stretch influenced remodeling of the extracellular matrix as well as cell-matrix interactions. These studies improve understanding of how vascular biology is affected by mechanical forces and how these molecules behave in cardiovascular diseases.
Collapse
Affiliation(s)
- T A Russo
- Department of Biochemistry, Molecular Biology Division, Carl Peter von Dietrich Laboratory, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil
| | - D Stoll
- Department of Nephrology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil
| | - H B Nader
- Department of Biochemistry, Molecular Biology Division, Carl Peter von Dietrich Laboratory, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil
| | - J L Dreyfuss
- Department of Biochemistry, Molecular Biology Division, Carl Peter von Dietrich Laboratory, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil..
| |
Collapse
|
23
|
Miyachi K, Yamada T, Kawagishi-Hotta M, Hasebe Y, Date Y, Hasegawa S, Arima M, Iwata Y, Kobayashi T, Numata S, Yamamoto N, Nakata S, Sugiura K, Akamatsu H. Extracellular proteoglycan decorin maintains human hair follicle stem cells. J Dermatol 2018; 45:1403-1410. [PMID: 30320452 DOI: 10.1111/1346-8138.14678] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 09/03/2018] [Indexed: 12/30/2022]
Abstract
Hair follicle stem cells (HFSC) are localized in the bulge region of the hair follicle and play a role in producing hair. Recently, it has been shown that the number of HFSC decreases with age, which is thought to be a cause of senile alopecia. Therefore, maintaining HFSC may be key for the prevention of age-related hair loss, but the regulatory mechanisms of HFSC and the effects of aging on them are largely unknown. In general, stem cells are known to require regulatory factors in the pericellular microenvironment, termed the stem cell niche, to maintain their cell function. In this study, we focused on the extracellular matrix proteoglycan decorin (DCN) as a candidate factor for maintaining the human HFSC niche. Gene expression analysis showed that DCN was highly expressed in the bulge region. We observed decreases in DCN expression as well as the number of KRT15-positive HFSC with age. In vitro experiments with human plucked hair-derived HFSC revealed that HFSC lost their undifferentiated state with increasing passages, and prior to this change a decrease in DCN expression was observed. Furthermore, knockdown of DCN promoted HFSC differentiation. In contrast, when HFSC were cultured on DCN-coated plates, they showed an even more undifferentiated state. From these results, as a novel mechanism for maintaining HFSC, it was suggested that DCN functions as a stem cell niche component, and that the deficit of HFSC maintenance caused by a reduction in DCN expression could be a cause of age-related hair loss.
Collapse
Affiliation(s)
- Katsuma Miyachi
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Nagoya, Japan
| | - Takaaki Yamada
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Nagoya, Japan.,Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Toyoake, Japan.,Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Mika Kawagishi-Hotta
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Nagoya, Japan.,Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Toyoake, Japan.,Nagoya University-MENARD Collaborative Chair, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Hasebe
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Nagoya, Japan.,Nagoya University-MENARD Collaborative Chair, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasushi Date
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Nagoya, Japan.,Nagoya University-MENARD Collaborative Chair, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiji Hasegawa
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Nagoya, Japan.,Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan.,Nagoya University-MENARD Collaborative Chair, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaru Arima
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yohei Iwata
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tsukane Kobayashi
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shigeki Numata
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Naoki Yamamoto
- Laboratory of Molecular Biology and Histochemistry, Joint Research Support Promotion Facility, Center for Research Promotion and Support, Fujita Health University, Toyoake, Japan
| | - Satoru Nakata
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Nagoya, Japan
| | - Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hirohiko Akamatsu
- Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
24
|
Extracellular Interactions between Fibulins and Transforming Growth Factor (TGF)-β in Physiological and Pathological Conditions. Int J Mol Sci 2018; 19:ijms19092787. [PMID: 30227601 PMCID: PMC6163299 DOI: 10.3390/ijms19092787] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/25/2022] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional peptide growth factor that has a vital role in the regulation of cell growth, differentiation, inflammation, and repair in a variety of tissues, and its dysregulation mediates a number of pathological conditions including fibrotic disorders, chronic inflammation, cardiovascular diseases, and cancer progression. Regulation of TGF-β signaling is multifold, but one critical site of regulation is via interaction with certain extracellular matrix (ECM) microenvironments, as TGF-β is primarily secreted as a biologically inactive form sequestrated into ECM. Several ECM proteins are known to modulate TGF-β signaling via cell–matrix interactions, including thrombospondins, SPARC (Secreted Protein Acidic and Rich in Cystein), tenascins, osteopontin, periostin, and fibulins. Fibulin family members consist of eight ECM glycoproteins characterized by a tandem array of calcium-binding epidermal growth factor-like modules and a common C-terminal domain. Fibulins not only participate in structural integrity of basement membrane and elastic fibers, but also serve as mediators for cellular processes and tissue remodeling as they are highly upregulated during embryonic development and certain disease processes, especially at the sites of epithelial–mesenchymal transition (EMT). Emerging studies have indicated a close relationship between fibulins and TGF-β signaling, but each fibulin plays a different role in a context-dependent manner. In this review, regulatory interactions between fibulins and TGF-β signaling are discussed. Understanding biological roles of fibulins in TGF-β regulation may introduce new insights into the pathogenesis of some human diseases.
Collapse
|
25
|
Tapella L, Cerruti M, Biocotino I, Stevano A, Rocchio F, Canonico PL, Grilli M, Genazzani AA, Lim D. TGF-β2 and TGF-β3 from cultured β-amyloid-treated or 3xTg-AD-derived astrocytes may mediate astrocyte-neuron communication. Eur J Neurosci 2018; 47:211-221. [DOI: 10.1111/ejn.13819] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Matteo Cerruti
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Isabella Biocotino
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Alessio Stevano
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Francesca Rocchio
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| |
Collapse
|
26
|
Wujak L, Schnieder J, Schaefer L, Wygrecka M. LRP1: A chameleon receptor of lung inflammation and repair. Matrix Biol 2017; 68-69:366-381. [PMID: 29262309 DOI: 10.1016/j.matbio.2017.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022]
Abstract
The lung displays a remarkable capability to regenerate following injury. Considerable effort has been made thus far to understand the cardinal processes underpinning inflammation and reconstruction of lung tissue. However, the factors determining the resolution or persistence of inflammation and efficient wound healing or aberrant remodeling remain largely unknown. Low density lipoprotein receptor-related protein 1 (LRP1) is an endocytic/signaling cell surface receptor which controls cellular and molecular mechanisms driving the physiological and pathological inflammatory reactions and tissue remodeling in several organs. In this review, we will discuss the impact of LRP1 on the consecutive steps of the inflammatory response and its role in the balanced tissue repair and aberrant remodeling in the lung.
Collapse
Affiliation(s)
- Lukasz Wujak
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Jennifer Schnieder
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Liliana Schaefer
- Goethe University School of Medicine, University Hospital, Theodor-Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany; Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
27
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
28
|
Ábrigo J, Campos F, Simon F, Riedel C, Cabrera D, Vilos C, Cabello-Verrugio C. TGF-β requires the activation of canonical and non-canonical signalling pathways to induce skeletal muscle atrophy. Biol Chem 2017; 399:253-264. [DOI: 10.1515/hsz-2017-0217] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/03/2017] [Indexed: 12/20/2022]
Abstract
Abstract
The transforming growth factor type-beta (TGF-β) induces skeletal muscle atrophy characterised by a decrease in the fibre’s diameter and levels of myosin heavy chain (MHC), also as an increase of MuRF-1 expression. In addition, TGF-β induces muscle atrophy by a mechanism dependent on reactive oxygen species (ROS). TGF-β signals by activating both canonical Smad-dependent, and non-canonical signalling pathways such as ERK1/2, JNK1/2, and p38 MAPKs. However, the participation of canonical and non-canonical signalling pathways in the TGF-β atrophic effect on skeletal muscle is unknown. We evaluate the impact of Smad and MAPK signalling pathways on the TGF-β-induced atrophic effect in C2C12 myotubes. The results indicate that TGF-β activates Smad2/3, ERK1/2 and JNK1/2, but not p38 in myotubes. The pharmacological inhibition of Smad3, ERK1/2 and JNK1/2 activation completely abolished the atrophic effect of TGF-β. Finally, the inhibition of these canonical and non-canonical pathways did not decrease the ROS increment, while the inhibition of ROS production entirely abolished the phosphorylation of Smad3, ERK1/2 and JNK1/2. These results suggest that TGF-β requires Smad3, ERK1/2 and JNK1/2 activation to produce skeletal muscle atrophy. Moreover, the induction of ROS by TGF-β is an upstream event to canonical and non-canonical pathways.
Collapse
Affiliation(s)
- Johanna Ábrigo
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Fabian Campos
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Felipe Simon
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Claudia Riedel
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Daniel Cabrera
- Universidad Bernardo O Higgins, Facultad de Salud , Departamento de Ciencias Químicas y Biológicas , 8370993 Santiago , Chile
- Departamento de Gastroenterología, Facultad de Medicina , Pontificia Universidad Católica de Chile , 8331150 Santiago , Chile
| | - Cristian Vilos
- Laboratory of Nanomedicine and Targeted Delivery, Center for Integrative Medicine and Innovative Science, Faculty of Medicine, and Center for Bioinformatics and Integrative Biology, Faculty of Biological Sciences , Universidad Andres Bello , 8370146 Santiago , Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA) , Universidad de Santiago de Chile , 9170022 Santiago , Chile
| | - Claudio Cabello-Verrugio
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
- Laboratory of Muscle Pathology, Fragility and Aging , Departmento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina , Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
| |
Collapse
|
29
|
Li Y, Hong J, Oh JE, Yoon AR, Yun CO. Potent antitumor effect of tumor microenvironment-targeted oncolytic adenovirus against desmoplastic pancreatic cancer. Int J Cancer 2017; 142:392-413. [PMID: 28929492 DOI: 10.1002/ijc.31060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/05/2017] [Indexed: 01/30/2023]
Abstract
Pancreatic cancer is a leading cause of cancer-related death. Desmoplastic pancreatic tumors exhibit excessive extracellular matrix (ECM) and are thus highly resistant to anticancer therapeutics, since the ECM restricts drug penetration and dispersion. Here, we designed and generated two hypoxia-responsive and cancer-specific hybrid promoters, H(mT)E and H(E)mT. Transgene expression driven by each hybrid promoter was markedly higher under hypoxic conditions than normoxic conditions. Moreover, H(E)mT-driven transgene expression was highly cancer-specific and was superior to that of H(mT)E-driven expression. A decorin-expressing oncolytic adenovirus (Ad; oH(E)mT-DCN) replicating under the control of the H(E)mT promoter induced more potent and highly cancer-specific cell death compared with its cognate control oncolytic Ad, which harbored the endogenous Ad E1A promoter. Moreover, oH(E)mT-DCN exhibited enhanced antitumor efficacy compared with both the clinically approved oncolytic Ad ONYX-015 and its cognate control oncolytic Ad lacking DCN. oH(E)mT-DCN treatment also attenuated the expression of major ECM components, such as collagen I/III, elastin and fibronectin and induced tumor cell apoptosis, leading to extensive viral dispersion within orthotopic pancreatic tumors and pancreatic cancer patient-derived tumor spheroids. Collectively, these findings demonstrate that oH(E)mT-DCN exhibits potent antitumor efficacy by degrading the ECM and inducing apoptosis in a multifunctional process. This process facilitates the dispersion and replication of oncolytic Ad, making it an attractive candidate for the treatment of aggressive and desmoplastic pancreatic cancer.
Collapse
Affiliation(s)
- Yan Li
- Graduate Program for Nanomedical Science, Yonsei University, Seoul, Korea
| | - JinWoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, Seongdong-Gu, Seoul, Korea
| | - Joung-Eun Oh
- Department of Bioengineering, College of Engineering, Hanyang University, Seongdong-Gu, Seoul, Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seongdong-Gu, Seoul, Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seongdong-Gu, Seoul, Korea
| |
Collapse
|
30
|
Sallustio F, Curci C, Aloisi A, Toma CC, Marulli E, Serino G, Cox SN, De Palma G, Stasi A, Divella C, Rinaldi R, Schena FP. Inhibin-A and Decorin Secreted by Human Adult Renal Stem/Progenitor Cells Through the TLR2 Engagement Induce Renal Tubular Cell Regeneration. Sci Rep 2017; 7:8225. [PMID: 28811645 PMCID: PMC5557965 DOI: 10.1038/s41598-017-08474-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
Abstract
Acute kidney injury (AKI) is a public health problem worldwide. Several therapeutic strategies have been made to accelerate recovery and improve renal survival. Recent studies have shown that human adult renal progenitor cells (ARPCs) participate in kidney repair processes, and may be used as a possible treatment to promote regeneration in acute kidney injury. Here, we show that human tubular ARPCs (tARPCs) protect physically injured or chemically damaged renal proximal tubular epithelial cells (RPTECs) by preventing cisplatin-induced apoptosis and enhancing proliferation of survived cells. tARPCs without toll-like receptor 2 (TLR2) expression or TLR2 blocking completely abrogated this regenerative effect. Only tARPCs, and not glomerular ARPCs, were able to induce tubular cell regeneration process and it occurred only after damage detection. Moreover, we have found that ARPCs secreted inhibin-A and decorin following the RPTEC damage and that these secreted factors were directly involved in cell regeneration process. Polysaccharide synthetic vesicles containing these molecules were constructed and co-cultured with cisplatin damaged RPTECs. These synthetic vesicles were not only incorporated into the cells, but they were also able to induce a substantial increase in cell number and viability. The findings of this study increase the knowledge of renal repair processes and may be the first step in the development of new specific therapeutic strategies for renal repair.
Collapse
Affiliation(s)
- Fabio Sallustio
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy.,C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy
| | - Claudia Curci
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy.,C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy.,Schena Foundation, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy
| | - Alessandra Aloisi
- Consiglio Nazionale delle Ricerche (CNR), Institute of Nanoscience, Via Arnesano 16, 73100, Lecce, Italy.,Institute of Microelectronics and Microsystems (C.N.R.- I.M.M.), via Monteroni, Campus Ecotekne, 73100, Lecce, Italy
| | - Chiara Cristina Toma
- Consiglio Nazionale delle Ricerche (CNR), Institute of Nanoscience, Via Arnesano 16, 73100, Lecce, Italy.,University of Salento, Mathematics and Physics "E. De Giorgi" Department, University of Salento, 73100, Lecce, Italy
| | - Elisabetta Marulli
- Consiglio Nazionale delle Ricerche (CNR), Institute of Nanoscience, Via Arnesano 16, 73100, Lecce, Italy.,University of Salento, Mathematics and Physics "E. De Giorgi" Department, University of Salento, 73100, Lecce, Italy
| | - Grazia Serino
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari, 70013, Italy
| | - Sharon Natasha Cox
- C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy.,Schena Foundation, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy
| | - Giuseppe De Palma
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy.,C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy.,Schena Foundation, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy
| | - Alessandra Stasi
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Chiara Divella
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Rosaria Rinaldi
- Consiglio Nazionale delle Ricerche (CNR), Institute of Nanoscience, Via Arnesano 16, 73100, Lecce, Italy.,University of Salento, Mathematics and Physics "E. De Giorgi" Department, University of Salento, 73100, Lecce, Italy
| | - Francesco Paolo Schena
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy. .,C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy. .,Schena Foundation, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy.
| |
Collapse
|
31
|
Revuelta-López E, Soler-Botija C, Nasarre L, Benitez-Amaro A, de Gonzalo-Calvo D, Bayes-Genis A, Llorente-Cortés V. Relationship among LRP1 expression, Pyk2 phosphorylation and MMP-9 activation in left ventricular remodelling after myocardial infarction. J Cell Mol Med 2017; 21:1915-1928. [PMID: 28378397 PMCID: PMC5571517 DOI: 10.1111/jcmm.13113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/30/2016] [Indexed: 01/09/2023] Open
Abstract
Left ventricular (LV) remodelling after myocardial infarction (MI) is a crucial determinant of the clinical course of heart failure. Matrix metalloproteinase (MMP) activation is strongly associated with LV remodelling after MI. Elucidation of plasma membrane receptors related to the activation of specific MMPs is fundamental for treating adverse cardiac remodelling after MI. The aim of current investigation was to explore the potential association between the low‐density lipoprotein receptor‐related protein 1 (LRP1) and MMP‐9 and MMP‐2 spatiotemporal expression after MI. Real‐time PCR and Western blot analyses showed that LRP1 mRNA and protein expression levels, respectively, were significantly increased in peri‐infarct and infarct zones at 10 and 21 days after MI. Confocal microscopy demonstrated high colocalization between LRP1 and the fibroblast marker vimentin, indicating that LRP1 is mostly expressed by cardiac fibroblasts in peri‐infarct and infarct areas. LRP1 also colocalized with proline‐rich tyrosine kinase 2 (pPyk2) and MMP‐9 in cardiac fibroblasts in ischaemic areas at 10 and 21 days after MI. Cell culture experiments revealed that hypoxia increases LRP1, pPyk2 protein levels and MMP‐9 activity in fibroblasts, without significant changes in MMP‐2 activity. MMP‐9 activation by hypoxia requires LRP1 and Pyk2 phosphorylation in fibroblasts. Collectively, our in vivo and in vitro data support a major role of cardiac fibroblast LRP1 levels on MMP‐9 up‐regulation associated with ventricular remodelling after MI.
Collapse
Affiliation(s)
- Elena Revuelta-López
- Cardiovascular Research Center, CSIC-ICCC, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,ICREC (Heart Failure and Cardiac Regeneration) Research Program, Health Sciences Research Institute Germans Tries i Pujol, Badalona (Barcelona), Spain
| | - Carol Soler-Botija
- ICREC (Heart Failure and Cardiac Regeneration) Research Program, Health Sciences Research Institute Germans Tries i Pujol, Badalona (Barcelona), Spain
| | - Laura Nasarre
- Cardiovascular Research Center, CSIC-ICCC, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Aleyda Benitez-Amaro
- Cardiovascular Research Center, CSIC-ICCC, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - David de Gonzalo-Calvo
- Cardiovascular Research Center, CSIC-ICCC, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC (Heart Failure and Cardiac Regeneration) Research Program, Health Sciences Research Institute Germans Tries i Pujol, Badalona (Barcelona), Spain.,Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vicenta Llorente-Cortés
- Cardiovascular Research Center, CSIC-ICCC, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
32
|
Fibulin-6 regulates pro-fibrotic TGF-β responses in neonatal mouse ventricular cardiac fibroblasts. Sci Rep 2017; 7:42725. [PMID: 28209981 PMCID: PMC5314373 DOI: 10.1038/srep42725] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/12/2017] [Indexed: 11/26/2022] Open
Abstract
Fibulin-6, an essential component of extracellular matrix determines the architecture of cellular junctions in tissues undergoing strain. Increased expression and deposition of fibulin-6 facilitates fibroblast migration in response to TGF-β, following myocardial infarction in mouse heart. The underlying mechanism still remains elusive. In conjunction with our previous study, we have now demonstrated that in fibulin-6 knockdown (KD) fibroblasts, not only TGF-β dependent migration, but also stress fiber formation, cellular networking and subsequently fibroblast wound contraction is almost abrogated. SMAD dependent TGF-β pathway shows ~75% decreased translocation of R-SMAD and co-SMAD into the nucleus upon fibulin-6 KD. Consequently, SMAD dependent pro-fibrotic gene expression is considerably down regulated to basal levels both in mRNA and protein. Also, investigating the non-SMAD pathways we observed a constitutive increase in pERK-levels in fibulin-6 KD fibroblast compared to control, but no change was seen in pAKT. Immunoprecipitation studies revealed 60% reduced interaction of TGF-β receptor II and I (TGFRII and I) accompanied by diminished phosphorylation of TGFRI at serin165 in fibulin-6 KD cells. In conclusion, fibulin-6 plays an important role in regulating TGF-β mediated responses, by modulating TGF-β receptor dimerization and activation to further trigger downstream pathways.
Collapse
|
33
|
Gubbiotti MA, Vallet SD, Ricard-Blum S, Iozzo RV. Decorin interacting network: A comprehensive analysis of decorin-binding partners and their versatile functions. Matrix Biol 2016; 55:7-21. [PMID: 27693454 DOI: 10.1016/j.matbio.2016.09.009] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Decorin, a prototype small leucine-rich proteoglycan, regulates a vast array of cellular processes including collagen fibrillogenesis, wound repair, angiostasis, tumor growth, and autophagy. This functional versatility arises from a wide array of decorin/protein interactions also including interactions with its single glycosaminoglycan side chain. The decorin-binding partners encompass numerous categories ranging from extracellular matrix molecules to cell surface receptors to growth factors and enzymes. Despite the diversity of the decorin interacting network, two main roles emerge as prominent themes in decorin function: maintenance of cellular structure and outside-in signaling, culminating in anti-tumorigenic effects. Here we present contemporary knowledge regarding the decorin interacting network and discuss in detail the biological relevance of these pleiotropic interactions, some of which could be targeted by therapeutic interventions.
Collapse
Affiliation(s)
- Maria A Gubbiotti
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sylvain D Vallet
- Pericellular and Extracellular Supramolecular Assemblies, Institute of Molecular and Supramolecular Chemistry and Biochemistry, University Claude Bernard, Lyon, France
| | - Sylvie Ricard-Blum
- Pericellular and Extracellular Supramolecular Assemblies, Institute of Molecular and Supramolecular Chemistry and Biochemistry, University Claude Bernard, Lyon, France
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
34
|
Goetsch KP, Niesler CU. The extracellular matrix regulates the effect of decorin and transforming growth factor beta-2 (TGF-β2) on myoblast migration. Biochem Biophys Res Commun 2016; 479:351-357. [PMID: 27644884 DOI: 10.1016/j.bbrc.2016.09.079] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/15/2016] [Indexed: 12/31/2022]
Abstract
Muscular injuries that destroy the basal lamina result in poor functional recovery of skeletal muscle. This is due, in part, to the deposition of structural fibrotic proteins such as fibronectin and collagen by fibroblasts and other cells. Transforming growth factor-β (TGF-β) promotes fibrosis, whereas the proteoglycan decorin is known to act as an anti-fibrotic agent, in part via the binding and neutralization of TGF-β. We have previously established that decorin can alter the migratory response of skeletal muscle myoblasts to the extracellular matrix (ECM) factor collagen, but not fibronectin. We have also shown that TGF-β reduces myoblast migration. In the current study we demonstrate that decorin can dramatically alter the inhibitory role of TGF-β on human myoblast migration and go on to shown that the extracellular matrix can significantly modify this effect. Decorin and TGF-β2 in combination were observed to significantly increase the rate of human myoblast migration, despite the inhibitory effect of TGF-β2 on its own. Furthermore, in the presence of fibronectin, TGF-β2 and decorin no longer acted synergistically to promote migration; while in the presence of collagen I, TGF-β2 failed to inhibit migration. These studies show, for the first time, that decorin can alter the bioactivity of TGF-β2 on human myoblast migration and emphasize the crucial regulatory role of the extracellular matrix in determining the response of skeletal muscle myoblasts to migratory cues.
Collapse
Affiliation(s)
- K P Goetsch
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - C U Niesler
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa.
| |
Collapse
|
35
|
Thu VT, Kim HK, Long LT, Thuy TT, Huy NQ, Kim SH, Kim N, Ko KS, Rhee BD, Han J. NecroX-5 exerts anti-inflammatory and anti-fibrotic effects via modulation of the TNFα/Dcn/TGFβ1/Smad2 pathway in hypoxia/reoxygenation-treated rat hearts. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:305-14. [PMID: 27162485 PMCID: PMC4860373 DOI: 10.4196/kjpp.2016.20.3.305] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 12/12/2022]
Abstract
Inflammatory and fibrotic responses are accelerated during the reperfusion period, and excessive fibrosis and inflammation contribute to cardiac malfunction. NecroX compounds have been shown to protect the liver and heart from ischemia-reperfusion injury. The aim of this study was to further define the role and mechanism of action of NecroX-5 in regulating infl ammation and fi brosis responses in a model of hypoxia/reoxygenation (HR). We utilized HR-treated rat hearts and lipopolysaccharide (LPS)-treated H9C2 culture cells in the presence or absence of NecroX-5 (10 µmol/L) treatment as experimental models. Addition of NecroX-5 signifi cantly increased decorin (Dcn) expression levels in HR-treated hearts. In contrast, expression of transforming growth factor beta 1 (TGFβ1) and Smad2 phosphorylation (pSmad2) was strongly attenuated in NecroX-5-treated hearts. In addition, signifi cantly increased production of tumor necrosis factor alpha (TNFα), TGFβ1, and pSmad2, and markedly decreased Dcn expression levels, were observed in LPS-stimulated H9C2 cells. Interestingly, NecroX-5 supplementation effectively attenuated the increased expression levels of TNFα, TGFβ1, and pSmad2, as well as the decreased expression of Dcn. Thus, our data demonstrate potential antiinflammatory and anti-fibrotic effects of NecroX-5 against cardiac HR injuries via modulation of the TNFα/Dcn/TGFβ1/Smad2 pathway.
Collapse
Affiliation(s)
- Vu Thi Thu
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.; VNU University of Science, Hanoi 120036, Vietnam
| | - Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.; Department of Integrated Biomedical Science, College of Medicine, Inje University, Busan 47392, Korea
| | - Le Thanh Long
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | | | | | - Soon Ha Kim
- Product Strategy and Development, LG Life Sciences Ltd., Seoul 03184, Korea
| | - Nari Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| |
Collapse
|
36
|
El Shafey N, Guesnon M, Simon F, Deprez E, Cosette J, Stockholm D, Scherman D, Bigey P, Kichler A. Inhibition of the myostatin/Smad signaling pathway by short decorin-derived peptides. Exp Cell Res 2016; 341:187-95. [PMID: 26844629 DOI: 10.1016/j.yexcr.2016.01.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/27/2016] [Accepted: 01/31/2016] [Indexed: 02/06/2023]
Abstract
Myostatin, also known as growth differentiation factor 8, is a member of the transforming growth factor-beta superfamily that has been shown to play a key role in the regulation of the skeletal muscle mass. Indeed, while myostatin deletion or loss of function induces muscle hypertrophy, its overexpression or systemic administration causes muscle atrophy. Since myostatin blockade is effective in increasing skeletal muscle mass, myostatin inhibitors have been actively sought after. Decorin, a member of the small leucine-rich proteoglycan family is a metalloprotein that was previously shown to bind and inactivate myostatin in a zinc-dependent manner. Furthermore, the myostatin-binding site has been shown to be located in the decorin N-terminal domain. In the present study, we investigated the anti-myostatin activity of short and soluble fragments of decorin. Our results indicate that the murine decorin peptides DCN48-71 and 42-65 are sufficient for inactivating myostatin in vitro. Moreover, we show that the interaction of mDCN48-71 to myostatin is strictly zinc-dependent. Binding of myostatin to activin type II receptor results in the phosphorylation of Smad2/3. Addition of the decorin peptide 48-71 decreased in a dose-dependent manner the myostatin-induced phosphorylation of Smad2 demonstrating thereby that the peptide inhibits the activation of the Smad signaling pathway. Finally, we found that mDCN48-71 displays a specificity towards myostatin, since it does not inhibit other members of the transforming growth factor-beta family.
Collapse
Affiliation(s)
- Nelly El Shafey
- Unité de Technologies Chimiques et Biologiques pour la Santé, CNRS UMR8258-Inserm, U1022 - Université Paris Descartes, Chimie ParisTech, 75006 Paris, France
| | - Mickaël Guesnon
- Unité de Technologies Chimiques et Biologiques pour la Santé, CNRS UMR8258-Inserm, U1022 - Université Paris Descartes, Chimie ParisTech, 75006 Paris, France
| | - Françoise Simon
- Laboratoire de Biologie et Pharmacologie Appliquée, ENS Cachan, UMR8113 CNRS, IDA FR3242, 94230 Cachan, France
| | - Eric Deprez
- Laboratoire de Biologie et Pharmacologie Appliquée, ENS Cachan, UMR8113 CNRS, IDA FR3242, 94230 Cachan, France
| | - Jérémie Cosette
- Inserm, UMR 951, Université d'Evry Val d'Essonne, Genethon, 91000 Evry, France
| | - Daniel Stockholm
- Inserm, UMR 951, Université d'Evry Val d'Essonne, Genethon, 91000 Evry, France
| | - Daniel Scherman
- Unité de Technologies Chimiques et Biologiques pour la Santé, CNRS UMR8258-Inserm, U1022 - Université Paris Descartes, Chimie ParisTech, 75006 Paris, France
| | - Pascal Bigey
- Unité de Technologies Chimiques et Biologiques pour la Santé, CNRS UMR8258-Inserm, U1022 - Université Paris Descartes, Chimie ParisTech, 75006 Paris, France
| | - Antoine Kichler
- Unité de Technologies Chimiques et Biologiques pour la Santé, CNRS UMR8258-Inserm, U1022 - Université Paris Descartes, Chimie ParisTech, 75006 Paris, France; Laboratoire de Conception et Application de Molécules Bioactives UMR7199 CNRS-Université de Strasbourg, LabEx Medalis, Faculté de Pharmacie, 67401 Illkirch, France.
| |
Collapse
|
37
|
Abrigo J, Rivera JC, Simon F, Cabrera D, Cabello-Verrugio C. Transforming growth factor type beta (TGF-β) requires reactive oxygen species to induce skeletal muscle atrophy. Cell Signal 2016; 28:366-376. [PMID: 26825874 DOI: 10.1016/j.cellsig.2016.01.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 12/28/2015] [Accepted: 01/24/2016] [Indexed: 12/13/2022]
Abstract
Transforming growth factor beta 1 (TGF-β1) is a classical modulator of skeletal muscle and regulates several processes, such as myogenesis, regeneration, and muscle function in skeletal muscle diseases. Skeletal muscle atrophy, characterised by the loss of muscle strength and mass, is one of the pathological conditions regulated by TGF-β. Atrophy also results in increased myosin heavy chain (MHC) degradation and the expression of two muscle-specific E3 ubiquitin ligases, atrogin-1 and MuRF-1. Reactive oxygen species (ROS) are modulators of muscle wasting, and NAD(P)H oxidase (NOX) is one of the main sources of ROS. While it was recently found that TGF-β1 induces atrophy in skeletal muscle, the underlying mechanism is not fully understood. In this study, the role of NOX-derived ROS in skeletal muscle atrophy induced by TGF-β was assessed. TGF-β1 induced an atrophic effect in C2C12 myotubes, as evidenced by decreased myotube diameter and MHC levels, together with increased MuRF-1 levels. Concomitantly, TGF-β increased NOX-induced ROS contents. Interestingly, NOX inhibition through apocynin and the antioxidant treatment with N-acetyl cysteine (NAC) decreased increased ROS levels in myotubes. Additionally, both apocynin and NAC completely prevented the decreased MHC, decreased myotube diameter, and increased MuRF-1 induced by TGF-β. Injection of TGF-β1 into the tibialis anterior muscle induced atrophy, as observed by decreased fibre diameter and MHC levels, together with increased MuRF-1 levels. Likewise, TGF-β increased the ROS contents in the smaller fibres of skeletal muscle. Additionally, the administration of NAC to mice prevented all atrophic effects and the increase in ROS induced by TGF-β in the tibialis anterior. This is the first study to report that TGF-β has an atrophic effect dependent on NOX-induced ROS in skeletal muscle.
Collapse
Affiliation(s)
- Johanna Abrigo
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Juan Carlos Rivera
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Laboratory of Integrative Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O Higgins, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| |
Collapse
|
38
|
Baghy K, Tátrai P, Regős E, Kovalszky I. Proteoglycans in liver cancer. World J Gastroenterol 2016; 22:379-393. [PMID: 26755884 PMCID: PMC4698501 DOI: 10.3748/wjg.v22.i1.379] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/14/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023] Open
Abstract
Proteoglycans are a group of molecules that contain at least one glycosaminoglycan chain, such as a heparan, dermatan, chondroitin, or keratan sulfate, covalently attached to the protein core. These molecules are categorized based on their structure, localization, and function, and can be found in the extracellular matrix, on the cell surface, and in the cytoplasm. Cell-surface heparan sulfate proteoglycans, such as syndecans, are the primary type present in healthy liver tissue. However, deterioration of the liver results in overproduction of other proteoglycan types. The purpose of this article is to provide a current summary of the most relevant data implicating proteoglycans in the development and progression of human and experimental liver cancer. A review of our work and other studies in the literature indicate that deterioration of liver function is accompanied by an increase in the amount of chondroitin sulfate proteoglycans. The alteration of proteoglycan composition interferes with the physiologic function of the liver on several levels. This article details and discusses the roles of syndecan-1, glypicans, agrin, perlecan, collagen XVIII/endostatin, endocan, serglycin, decorin, biglycan, asporin, fibromodulin, lumican, and versican in liver function. Specifically, glypicans, agrin, and versican play significant roles in the development of liver cancer. Conversely, the presence of decorin could potentially provide protective effects.
Collapse
|
39
|
Decorin: A Growth Factor Antagonist for Tumor Growth Inhibition. BIOMED RESEARCH INTERNATIONAL 2015; 2015:654765. [PMID: 26697491 PMCID: PMC4677162 DOI: 10.1155/2015/654765] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/21/2015] [Indexed: 01/12/2023]
Abstract
Decorin (DCN) is the best characterized member of the extracellular small leucine-rich proteoglycan family present in connective tissues, typically in association with or “decorating” collagen fibrils. It has substantial interest to clinical medicine owing to its antifibrotic, anti-inflammatory, and anticancer effects. Studies on DCN knockout mice have established that a lack of DCN is permissive for tumor development and it is regarded as a tumor suppressor gene. A reduced expression or a total disappearance of DCN has been reported to take place in various forms of human cancers during tumor progression. Furthermore, when used as a therapeutic molecule, DCN has been shown to inhibit tumor progression and metastases in experimental cancer models. DCN affects the biology of various types of cancer by targeting a number of crucial signaling molecules involved in cell growth, survival, metastasis, and angiogenesis. The active sites for the neutralization of different growth factors all reside in different parts of the DCN molecule. An emerging concept that multiple proteases, especially those produced by inflammatory cells, are capable of cleaving DCN suggests that native DCN could be inactivated in a number of pathological inflammatory conditions. In this paper, we review the role of DCN in cancer.
Collapse
|
40
|
Cohen DPA, Martignetti L, Robine S, Barillot E, Zinovyev A, Calzone L. Mathematical Modelling of Molecular Pathways Enabling Tumour Cell Invasion and Migration. PLoS Comput Biol 2015; 11:e1004571. [PMID: 26528548 PMCID: PMC4631357 DOI: 10.1371/journal.pcbi.1004571] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 09/29/2015] [Indexed: 02/07/2023] Open
Abstract
Understanding the etiology of metastasis is very important in clinical perspective, since it is estimated that metastasis accounts for 90% of cancer patient mortality. Metastasis results from a sequence of multiple steps including invasion and migration. The early stages of metastasis are tightly controlled in normal cells and can be drastically affected by malignant mutations; therefore, they might constitute the principal determinants of the overall metastatic rate even if the later stages take long to occur. To elucidate the role of individual mutations or their combinations affecting the metastatic development, a logical model has been constructed that recapitulates published experimental results of known gene perturbations on local invasion and migration processes, and predict the effect of not yet experimentally assessed mutations. The model has been validated using experimental data on transcriptome dynamics following TGF-β-dependent induction of Epithelial to Mesenchymal Transition in lung cancer cell lines. A method to associate gene expression profiles with different stable state solutions of the logical model has been developed for that purpose. In addition, we have systematically predicted alleviating (masking) and synergistic pairwise genetic interactions between the genes composing the model with respect to the probability of acquiring the metastatic phenotype. We focused on several unexpected synergistic genetic interactions leading to theoretically very high metastasis probability. Among them, the synergistic combination of Notch overexpression and p53 deletion shows one of the strongest effects, which is in agreement with a recent published experiment in a mouse model of gut cancer. The mathematical model can recapitulate experimental mutations in both cell line and mouse models. Furthermore, the model predicts new gene perturbations that affect the early steps of metastasis underlying potential intervention points for innovative therapeutic strategies in oncology.
Collapse
Affiliation(s)
- David P. A. Cohen
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Loredana Martignetti
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Sylvie Robine
- Institut Curie, Paris, France
- CNRS UMR144, Paris, France
| | - Emmanuel Barillot
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Andrei Zinovyev
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Laurence Calzone
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
- * E-mail:
| |
Collapse
|
41
|
Abrigo J, Morales MG, Simon F, Cabrera D, Di Capua G, Cabello-Verrugio C. Apocynin inhibits the upregulation of TGF-β1 expression and ROS production induced by TGF-β in skeletal muscle cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:885-893. [PMID: 26321737 DOI: 10.1016/j.phymed.2015.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/18/2015] [Accepted: 06/21/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Pure apocynin, which can be traditionally isolated and purified from several plant species such as Picrorhiza kurroa Royle ex Benth (Scrophulariaceae), acts as an inhibitor of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activity inhibiting its production of reactive oxygen species (ROS). Transforming growth factor type beta 1 (TGF-β1) is a growth factor that produces inhibition of myogenesis, diminution of regeneration and induction of atrophy in skeletal muscle. The typical signalling that is activated by TGF-β involves the Smad pathway. PURPOSE To evaluate the effect of TGF-β and the effect of apocynin on TGF-β1 expression in skeletal muscle cells. STUDY DESIGN Controlled laboratory study. In vitro assays were performed with C2C12 cells incubated with TGF-β1 in presence or absence of apocynin (NOX inhibitor), SB525334 (TGF-β-receptor I inhibitor), or chelerythrine (PKC inhibitor). METHODS TGF-β1 and atrogin-1 expression was evaluated by RT-qPCR and/or ELISA; Smad3 phosphorylation by western blot; Smad4 nuclear translocation by indirect immunofluorescence; and ROS levels by DCF probe fluorescent measurements. RESULTS We show that myoblasts respond to TGF-β1 by increasing its own gene expression in a time- and dose-dependent fashion which was abolished by SB525334 and siRNA for Smad2/3. TGF-β1 also induced ROS. Remarkably, apocynin inhibited the TGF-β1 induced ROS as well as the autoinduction of TGF-β1 gene expression. We also show that TGF-β-induced ROS production and TGF-β1 expression require PKC activity as indicated by the inhibition using chelerythrine. CONCLUSION These results strongly suggest that TGF-β induces its own expression through a TGF-β-receptor/Smad-dependent mechanism and apocynin is able to inhibit this process, suggesting that requires NOX-induced ROS in skeletal muscle cells.
Collapse
Affiliation(s)
- Johanna Abrigo
- Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - María Gabriela Morales
- Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Simon
- Laboratorio de Fisiopatología Integrativa, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Daniel Cabrera
- Departamento de Ciencias Químicas y Biológicas, Universidad Bernardo O´Higgins, Santiago, Chile
| | - Gabriella Di Capua
- Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| |
Collapse
|
42
|
Neill T, Schaefer L, Iozzo RV. Decoding the Matrix: Instructive Roles of Proteoglycan Receptors. Biochemistry 2015; 54:4583-98. [PMID: 26177309 DOI: 10.1021/acs.biochem.5b00653] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The extracellular matrix is a dynamic repository harboring instructive cues that embody substantial regulatory dominance over many evolutionarily conserved intracellular activities, including proliferation, apoptosis, migration, motility, and autophagy. The matrix also coordinates and parses hierarchical information, such as angiogenesis, tumorigenesis, and immunological responses, typically providing the critical determinants driving each outcome. We provide the first comprehensive review focused on proteoglycan receptors, that is, signaling transmembrane proteins that use secreted proteoglycans as ligands, in addition to their natural ligands. The majority of these receptors belong to an exclusive subset of receptor tyrosine kinases and assorted cell surface receptors that specifically bind, transduce, and modulate fundamental cellular processes following interactions with proteoglycans. The class of small leucine-rich proteoglycans is the most studied so far and constitutes the best understood example of proteoglycan-receptor interactions. Decorin and biglycan evoke autophagy and immunological responses that deter, suppress, or exacerbate pathological conditions such as tumorigenesis, angiogenesis, and chronic inflammatory disease. Basement membrane-associated heparan sulfate proteoglycans (perlecan, agrin, and collagen XVIII) represent a unique cohort and provide proteolytically cleaved bioactive fragments for modulating cellular behavior. The receptors that bind the genuinely multifactorial and multivalent proteoglycans represent a nexus in understanding basic biological pathways and open new avenues for therapeutic and pharmacological intervention.
Collapse
Affiliation(s)
- Thomas Neill
- †Department of Pathology, Anatomy and Cell Biology and Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Liliana Schaefer
- ‡Department of Pharmacology, Goethe University, 60590 Frankfurt, Germany
| | - Renato V Iozzo
- †Department of Pathology, Anatomy and Cell Biology and Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| |
Collapse
|
43
|
Le Bihan MC, Barrio-Hernandez I, Mortensen TP, Henningsen J, Jensen SS, Bigot A, Blagoev B, Butler-Browne G, Kratchmarova I. Cellular Proteome Dynamics during Differentiation of Human Primary Myoblasts. J Proteome Res 2015; 14:3348-61. [DOI: 10.1021/acs.jproteome.5b00397] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Marie-Catherine Le Bihan
- Department
of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Inigo Barrio-Hernandez
- Department
of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Tenna Pavia Mortensen
- Department
of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Jeanette Henningsen
- Department
of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Søren Skov Jensen
- Department
of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Anne Bigot
- Center
for Research in Myology, Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS975, CNRS FRE3617, 75013 Paris, France
| | - Blagoy Blagoev
- Department
of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Gillian Butler-Browne
- Center
for Research in Myology, Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS975, CNRS FRE3617, 75013 Paris, France
| | - Irina Kratchmarova
- Department
of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| |
Collapse
|
44
|
Radeke MJ, Radeke CM, Shih YH, Hu J, Bok D, Johnson LV, Coffey PJ. Restoration of mesenchymal retinal pigmented epithelial cells by TGFβ pathway inhibitors: implications for age-related macular degeneration. Genome Med 2015; 7:58. [PMID: 26150894 PMCID: PMC4491894 DOI: 10.1186/s13073-015-0183-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 06/11/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD) is a leading cause of blindness. Most vision loss occurs following the transition from a disease of deposit formation and inflammation to a disease of neovascular fibrosis and/or cell death. Here, we investigate how repeated wound stimulus leads to seminal changes in gene expression and the onset of a perpetual state of stimulus-independent wound response in retinal pigmented epithelial (RPE) cells, a cell-type central to the etiology of AMD. METHODS Transcriptome wide expression profiles of human fetal RPE cell cultures as a function of passage and time post-plating were determined using Agilent 44 K whole genome microarrays and RNA-Seq. Using a systems level analysis, differentially expressed genes and pathways of interest were identified and their role in the establishment of a persistent mesenchymal state was assessed using pharmacological-based experiments. RESULTS Using a human fetal RPE cell culture model that considers monolayer disruption and subconfluent culture as a proxy for wound stimulus, we show that prolonged wound stimulus leads to terminal acquisition of a mesenchymal phenotype post-confluence and altered expression of more than 40 % of the transcriptome. In contrast, at subconfluence fewer than 5 % of expressed transcripts have two-fold or greater expression differences after repeated passage. Protein-protein and pathway interaction analysis of the genes with passage-dependent expression levels in subconfluent cultures reveals a 158-node interactome comprised of two interconnected modules with functions pertaining to wound response and cell division. Among the wound response genes are the TGFβ pathway activators: TGFB1, TGFB2, INHBA, INHBB, GDF6, CTGF, and THBS1. Significantly, inhibition of TGFBR1/ACVR1B mediated signaling using receptor kinase inhibitors both forestalls and largely reverses the passage-dependent loss of epithelial potential; thus extending the effective lifespan by at least four passages. Moreover, a disproportionate number of RPE wound response genes have altered expression in neovascular and geographic AMD, including key members of the TGFβ pathway. CONCLUSIONS In RPE cells the switch to a persistent mesenchymal state following prolonged wound stimulus is driven by lasting activation of the TGFβ pathway. Targeted inhibition of TGFβ signaling may be an effective approach towards retarding AMD progression and producing RPE cells in quantity for research and cell-based therapies.
Collapse
Affiliation(s)
- Monte J. Radeke
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| | - Carolyn M. Radeke
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| | - Ying-Hsuan Shih
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| | - Jane Hu
- />Departments of Ophthalmology and Neurobiology, Jules Stein Eye & Brain Research Institutes, David Geffen School of Medicine, University of California, Los Angeles, CA USA
| | - Dean Bok
- />Departments of Ophthalmology and Neurobiology, Jules Stein Eye & Brain Research Institutes, David Geffen School of Medicine, University of California, Los Angeles, CA USA
| | - Lincoln V. Johnson
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| | - Pete J. Coffey
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| |
Collapse
|
45
|
Nishimura T. Role of extracellular matrix in development of skeletal muscle and postmortem aging of meat. Meat Sci 2015; 109:48-55. [PMID: 26141816 DOI: 10.1016/j.meatsci.2015.05.015] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 05/15/2015] [Accepted: 05/16/2015] [Indexed: 01/05/2023]
Abstract
The integrity of skeletal muscle is maintained by the intramuscular connective tissues (IMCTs) that are composed of extracellular matrix (ECM) molecules such as collagens, proteoglycans, and glycoproteins. The ECM plays an important role not only in providing biomechanical strength of the IMCT, but also in regulating muscle cell behavior. Some ECM molecules, such as decorin and laminin, modulate the activity of myostatin that regulates skeletal muscle mass. Furthermore, it has been shown that decorin activates Akt downstream of insulin-like growth factor-I receptor (IGF-IR) and enhances the differentiation of myogenic cells, suggesting that decorin acts as a signaling molecule to myogenic cells. With animal growth, the structural integrity of IMCT increases; collagen fibrils within the endomysium associate more closely with each other, and the collagen fibers in the perimysium become increasingly thick and their wavy pattern grows more regular. These changes increase the mechanical strength of IMCT, contributing to the toughening of meat. However, in highly marbled beef cattle like Wagyu, intramuscular fat deposits mainly in the perimysium between muscle fiber bundles during the fattening period. The development of adipose tissues appears to disorganize the structure of IMCT and contributes to the tenderness of Wagyu beef. The IMCT was considered to be rather immutable compared to myofibrils during postmortem aging of meat. However, several studies have shown that collagen networks in the IMCT are disintegrated and proteoglycan components are degraded during postmortem aging. These changes in ECM appear to reduce the mechanical strength of IMCT and contribute to the tenderness of uncooked meat or cooked meat at low temperature. Thus, the ECM plays a multifunctional role in skeletal muscle development and postmortem aging of meat.
Collapse
Affiliation(s)
- Takanori Nishimura
- Muscle Biology and Meat Science Laboratory, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
46
|
Cofre C, Acuña MJ, Contreras O, Morales MG, Riquelme C, Cabello-Verrugio C, Brandan E. Transforming growth factor type-β inhibits Mas receptor expression in fibroblasts but not in myoblasts or differentiated myotubes; Relevance to fibrosis associated to muscular dystrophies. Biofactors 2015; 41:111-20. [PMID: 25809912 DOI: 10.1002/biof.1208] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/26/2015] [Indexed: 12/28/2022]
Abstract
Duchenne muscular dystrophy is a genetic disorder characterized by myofiber degeneration, muscle weakness, and increased fibrosis. Transforming growth factor type-β (TGF-β), a central mediator of fibrosis, is upregulated in fibrotic diseases. Angiotensin-(1-7) [Ang-(1-7)] is a peptide with actions that oppose those of angiotensin-II (Ang II). Ang-(1-7) effects are mediated by the Mas receptor. Treatment with Ang-(1-7) produce positive effects in the mdx mouse, normalizing skeletal muscle architecture, decreasing local fibrosis, and fibroblasts, and improving muscle function. Mdx mice deficient for the Mas receptor showed the opposite effects. To identify the cell type(s) responsible for Mas receptor expression, and to characterize whether profibrotic effectors had any effect on its expression, we determined the effect of profibrotic agents on Mas expression. TGF-β, but not connective tissue growth factor or Ang-II, reduced the expression of Mas receptor in fibroblasts isolated from skeletal muscle cells and fibroblasts from two established cell lines. In contrast, no effects were observed in myoblasts and differentiated myotubes. This inhibition was mediated by the Smad-dependent (canonical) and the PI3K and MEK1/2 (noncanonical) TGF-β signaling pathways. When both canonical and noncanonical inhibitors of the TGF-β-dependent pathways were added together, the inhibitory effect of TGF-β on Mas expression was lost. The decrease in Mas receptor induced by TGF-β in fibroblasts reduced the Ang-(1-7) mediated stimulation of phosphorylation of AKT pathway proteins. These results suggest that reduction of Mas receptor in fibroblasts, by TGF-β, could increase the fibrotic phenotype observed in dystrophic skeletal muscle decreasing the beneficial effect of Ang-(1-7).
Collapse
MESH Headings
- Angiotensin I/pharmacology
- Angiotensin II/pharmacology
- Animals
- Cell Line
- Disease Models, Animal
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Fibrosis
- Gene Expression Regulation
- MAP Kinase Kinase 1/genetics
- MAP Kinase Kinase 1/metabolism
- MAP Kinase Kinase 2/genetics
- MAP Kinase Kinase 2/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Knockout
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myoblasts/drug effects
- Myoblasts/metabolism
- Myoblasts/pathology
- Organ Specificity
- Peptide Fragments/pharmacology
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation/drug effects
- Primary Cell Culture
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- Catalina Cofre
- Center for Aging and Regeneration, CARE Chile-UC and Department of Cell and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
47
|
Iozzo RV, Schaefer L. Proteoglycan form and function: A comprehensive nomenclature of proteoglycans. Matrix Biol 2015; 42:11-55. [PMID: 25701227 PMCID: PMC4859157 DOI: 10.1016/j.matbio.2015.02.003] [Citation(s) in RCA: 804] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
We provide a comprehensive classification of the proteoglycan gene families and respective protein cores. This updated nomenclature is based on three criteria: Cellular and subcellular location, overall gene/protein homology, and the utilization of specific protein modules within their respective protein cores. These three signatures were utilized to design four major classes of proteoglycans with distinct forms and functions: the intracellular, cell-surface, pericellular and extracellular proteoglycans. The proposed nomenclature encompasses forty-three distinct proteoglycan-encoding genes and many alternatively-spliced variants. The biological functions of these four proteoglycan families are critically assessed in development, cancer and angiogenesis, and in various acquired and genetic diseases where their expression is aberrant.
Collapse
Affiliation(s)
- Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany.
| |
Collapse
|
48
|
Ding Q, Zhang M, Liu C. Asporin participates in gastric cancer cell growth and migration by influencing EGF receptor signaling. Oncol Rep 2015; 33:1783-90. [PMID: 25673058 DOI: 10.3892/or.2015.3791] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 01/16/2015] [Indexed: 11/06/2022] Open
Abstract
Asporin (ASPN), a novel member of the small leucine-rich proteoglycan (SLRP) family, serves as a key component of the tumor stroma and has been reported to be abnormally expressed in certain types of tumors. Specifically, the proteoglycan was proven to activate the coordinated invasion of scirrhous gastric cancer and cancer-associated fibroblasts. However, the role of ASPN in cancer cell growth and metastasis has not yet been addressed. In the present study, we aimed to evaluate the tumoricidal benefits of ASPN on tumorigenesis and progression of gastric cancer. Firstly, it was demonstrated that ASPN was overexpressed in gastric carcinoma tissues when compared to the corresponding non‑cancerous tissues, and it had varied levels of expression in gastric cancer epithelial cell lines. Additionally, we assessed the effects of transient siRNA‑mediated ASPN knockdown on gastric cancer cells. ASPN silencing inhibited proliferation and suppressed the migration of immortalized neoplastic epithelial cells. Furthermore, at the molecular level, we found that downregulation of ASPN blocked the anti-apoptotic molecule Bcl-2, increased the expression of pro-apoptotic molecule Bad, reduced the expression of migration-related proteins CD44 and matrix metalloproteinase (MMP)-2, and abrogated the activation of the phosphorylation status of ERK and epidermal growth factor (EGF) and its receptor (EGFR). Collectively, our findings indicate that ASPN is upregulated and plays an oncogenic role in gastric cancer progression and metastasis by influencing the EGFR signaling pathway.
Collapse
Affiliation(s)
- Qian Ding
- Department of Ultrasound, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Mei Zhang
- Department of Ultrasound, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Can Liu
- Department of Ultrasound, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
49
|
Lin L, Hu K. LRP-1: functions, signaling and implications in kidney and other diseases. Int J Mol Sci 2014; 15:22887-901. [PMID: 25514242 PMCID: PMC4284744 DOI: 10.3390/ijms151222887] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/06/2014] [Accepted: 12/04/2014] [Indexed: 12/17/2022] Open
Abstract
Low-density lipoprotein (LDL)-related protein-1 (LRP-1) is a member of LDL receptor family that is implicated in lipoprotein metabolism and in the homeostasis of proteases and protease inhibitors. Expression of LRP-1 is ubiquitous. Up-regulation of LRP-1 has been reported in numerous human diseases. In addition to its function as a scavenger receptor for various ligands, LRP-1 has been shown to transduce multiple intracellular signal pathways including mitogen-activated protein kinase (MAPK), Akt, Rho, and the integrin signaling. LRP-1 signaling plays an important role in the regulation of diverse cellular process, such as cell proliferation, survival, motility, differentiation, and transdifferentiation, and thus participates in the pathogenesis of organ dysfunction and injury. In this review, we focus on the current understanding of LRP-1 signaling and its roles in the development and progression of kidney disease. The role and signaling of LRP-1 in the nervous and cardiovascular systems, as well as in carcinogenesis, are also briefly discussed.
Collapse
Affiliation(s)
- Ling Lin
- Division of Nephrology, Department of Medicine, College of Medicine, Penn State University, 500 University Drive, Hershey, PA 17033, USA.
| | - Kebin Hu
- Division of Nephrology, Department of Medicine, College of Medicine, Penn State University, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
50
|
Gene expression profiling of giant cell tumor of bone reveals downregulation of extracellular matrix components decorin and lumican associated with lung metastasis. Virchows Arch 2014; 465:703-13. [DOI: 10.1007/s00428-014-1666-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 08/21/2014] [Accepted: 10/03/2014] [Indexed: 11/26/2022]
|