1
|
Yamamoto K, Scilabra SD, Bonelli S, Jensen A, Scavenius C, Enghild JJ, Strickland DK. Novel insights into the multifaceted and tissue-specific roles of the endocytic receptor LRP1. J Biol Chem 2024; 300:107521. [PMID: 38950861 PMCID: PMC11325810 DOI: 10.1016/j.jbc.2024.107521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Receptor-mediated endocytosis provides a mechanism for the selective uptake of specific molecules thereby controlling the composition of the extracellular environment and biological processes. The low-density lipoprotein receptor-related protein 1 (LRP1) is a widely expressed endocytic receptor that regulates cellular events by modulating the levels of numerous extracellular molecules via rapid endocytic removal. LRP1 also participates in signalling pathways through this modulation as well as in the interaction with membrane receptors and cytoplasmic adaptor proteins. LRP1 SNPs are associated with several diseases and conditions such as migraines, aortic aneurysms, cardiopulmonary dysfunction, corneal clouding, and bone dysmorphology and mineral density. Studies using Lrp1 KO mice revealed a critical, nonredundant and tissue-specific role of LRP1 in regulating various physiological events. However, exactly how LRP1 functions to regulate so many distinct and specific processes is still not fully clear. Our recent proteomics studies have identified more than 300 secreted proteins that either directly interact with LRP1 or are modulated by LRP1 in various tissues. This review will highlight the remarkable ability of this receptor to regulate secreted molecules in a tissue-specific manner and discuss potential mechanisms underpinning such specificity. Uncovering the depth of these "hidden" specific interactions modulated by LRP1 will provide novel insights into a dynamic and complex extracellular environment that is involved in diverse biological and pathological processes.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Simone D Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy
| | - Simone Bonelli
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Zhang N, Wu X, Zhang W, Sun Y, Yan X, Xu A, Han Q, Yang A, You H, Chen W. Targeting thrombospondin-2 retards liver fibrosis by inhibiting TLR4-FAK/TGF-β signaling. JHEP Rep 2024; 6:101014. [PMID: 38379585 PMCID: PMC10877131 DOI: 10.1016/j.jhepr.2024.101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 02/22/2024] Open
Abstract
Background & Aims Thrombospondin-2 (THBS2) expression is associated with liver fibrosis regardless of etiology. However, the role of THBS2 in the pathogenesis of liver fibrosis has yet to be elucidated. Methods The in vivo effects of silencing Thbs2 in hepatic stellate cells (HSCs) were examined using an adeno-associated virus vector (serotype 6, AAV6) containing short-hairpin RNAs targeting Thbs2, under the regulatory control of cytomegalovirus, U6 or the α-smooth muscle promoter, in mouse models of carbon tetrachloride or methionine-choline deficient (MCD) diet-induced liver fibrosis. Crosstalk between THBS2 and toll-like receptor 4 (TLR4), as well as the cascaded signaling, was systematically investigated using mouse models, primary HSCs, and human HSC cell lines. Results THBS2 was predominantly expressed in activated HSCs and dynamically increased with liver fibrosis progression and decreased with regression. Selective interference of Thbs2 in HSCs retarded intrahepatic inflammatory infiltration, steatosis accumulation, and fibrosis progression following carbon tetrachloride challenge or in a dietary model of metabolic dysfunction-associated steatohepatitis. Mechanically, extracellular THBS2, as a dimer, specifically recognized and directly bound to TLR4, activating HSCs by stimulating downstream profibrotic focal adhesion kinase (FAK)/transforming growth factor beta (TGF-β) pathways. Disruption of the THBS2-TLR4-FAK/TGF-β signaling axis notably alleviated HSC activation and liver fibrosis aggravation. Conclusions THBS2 plays a crucial role in HSC activation and liver fibrosis progression through TLR4-FAK/TGF-β signaling in an autocrine manner, representing an attractive potential therapeutic target for liver fibrosis. Impact and implications Thrombospondin-2 (THBS2) is emerging as a factor closely associated with liver fibrosis regardless of etiology. However, the mechanisms by which THBS2 is involved in liver fibrosis remain unclear. Here, we showed that THBS2 plays a prominent role in the pathogenesis of liver fibrosis by activating the TLR4-TGF-β/FAK signaling axis and hepatic stellate cells in an autocrine manner, providing a potential therapeutic target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ning Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Xiaoning Wu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Wen Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Yameng Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Xuzhen Yan
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Anjian Xu
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Qi Han
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Aiting Yang
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Wei Chen
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| |
Collapse
|
3
|
Arrigo AB, Zhu W, Williams KA, Guzman-Moreno C, Lo C, Lin JHI. Contribution of LRP1 in Human Congenital Heart Disease Correlates with Its Roles in the Outflow Tract and Atrioventricular Cushion Development. Genes (Basel) 2023; 14:genes14040947. [PMID: 37107705 PMCID: PMC10137934 DOI: 10.3390/genes14040947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Due to the prevalence of congenital heart disease in the human population, determining the role of variants in congenital heart disease (CHD) can give a better understanding of the cause of the disorder. A homozygous missense mutation in the LDL receptor-related protein 1 (Lrp1) in mice was shown to cause congenital heart defects, including atrioventricular septal defect (AVSD) and double outlet right ventricle (DORV). Integrative analysis of publicly available single-cell RNA sequencing (scRNA-seq) datasets and spatial transcriptomics of human and mouse hearts indicated that LRP1 is predominantly expressed in mesenchymal cells and mainly located in the developing outflow tract and atrioventricular cushion. Gene burden analysis of 1922 CHD individuals versus 2602 controls with whole-exome sequencing showed a significant excess of rare damaging LRP1 mutations in CHD (odds ratio (OR) = 2.22, p = 1.92 × 10-4), especially in conotruncal defect with OR of 2.37 (p = 1.77 × 10-3) and atrioventricular septal defect with OR of 3.14 (p = 0.0194). Interestingly, there is a significant relationship between those variants that have an allele frequency below 0.01% and atrioventricular septal defect, which is the phenotype observed previously in a homozygous N-ethyl-N-nitrosourea (ENU)-induced Lrp1 mutant mouse line.
Collapse
Affiliation(s)
- Angelo B Arrigo
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
| | - Wenjuan Zhu
- Centre for Cardiovascular Genomics and Medicine, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Kylia A Williams
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
| | - Carla Guzman-Moreno
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
| | - Cecilia Lo
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
| | - Jiuann-Huey I Lin
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, 530 45th St, Pittsburgh, PA 15201, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
- UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA
| |
Collapse
|
4
|
A midposition NOTCH3 truncation in inherited cerebral small vessel disease may affect the protein interactome. J Biol Chem 2022; 299:102772. [PMID: 36470429 PMCID: PMC9808000 DOI: 10.1016/j.jbc.2022.102772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 12/07/2022] Open
Abstract
Mutations in NOTCH3 underlie cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), the most common inherited cerebral small vessel disease. Two cleavages of NOTCH3 protein, at Asp80 and Asp121, were previously described in CADASIL pathological samples. Using monoclonal antibodies developed against a NOTCH3 neoepitope, we identified a third cleavage at Asp964 between an Asp-Pro sequence. We characterized the structural requirements for proteolysis at Asp964 and the vascular distribution of the cleavage event. A proteome-wide analysis was performed to find proteins that interact with the cleavage product. Finally, we investigated the biochemical determinants of this third cleavage event. Cleavage at Asp964 was critically dependent on the proline adjacent to the aspartate residue. In addition, the cleavage product was highly enriched in CADASIL brain tissue and localized to the media of degenerating arteries, where it deposited with the two additional NOTCH3 cleavage products. Recombinant NOTCH3 terminating at Asp964 was used to probe protein microarrays. We identified multiple molecules that bound to the cleaved NOTCH3 more than to uncleaved protein, suggesting that cleavage may alter the local protein interactome within disease-affected blood vessels. The cleavage of purified NOTCH3 protein at Asp964 in vitro was activated by reducing agents and NOTCH3 protein; cleavage was inhibited by specific dicarboxylic acids, as seen with cleavage at Asp80 and Asp121. Overall, we propose homologous redox-driven Asp-Pro cleavages and alterations in protein interactions as potential mechanisms in inherited small vessel disease; similarities in protein cleavage characteristics may indicate common biochemical modulators of pathological NOTCH3 processing.
Collapse
|
5
|
Friedrich T, Ferrante F, Pioger L, Nist A, Stiewe T, Andrau JC, Bartkuhn M, Giaimo BD, Borggrefe T. Notch-dependent and -independent functions of transcription factor RBPJ. Nucleic Acids Res 2022; 50:7925-7937. [PMID: 35848919 PMCID: PMC9371899 DOI: 10.1093/nar/gkac601] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/27/2022] [Accepted: 07/05/2022] [Indexed: 11/14/2022] Open
Abstract
Signal transduction pathways often involve transcription factors that promote activation of defined target gene sets. The transcription factor RBPJ is the central player in Notch signaling and either forms an activator complex with the Notch intracellular domain (NICD) or a repressor complex with corepressors like KYOT2/FHL1. The balance between these two antagonizing RBPJ-complexes depends on the activation state of the Notch receptor regulated by cell-to-cell interaction, ligand binding and proteolytic cleavage events. Here, we depleted RBPJ in mature T-cells lacking active Notch signaling and performed RNA-Seq, ChIP-Seq and ATAC-seq analyses. RBPJ depletion leads to upregulation of many Notch target genes. Ectopic expression of NICD1 activates several Notch target genes and enhances RBPJ occupancy. Based on gene expression changes and RBPJ occupancy we define four different clusters, either RBPJ- and/or Notch-regulated genes. Importantly, we identify early (Hes1 and Hey1) and late Notch-responsive genes (IL2ra). Similarly, to RBPJ depletion, interfering with transcriptional repression by squelching with cofactor KYOT2/FHL1, leads to upregulation of Notch target genes. Taken together, RBPJ is not only an essential part of the Notch co-activator complex but also functions as a repressor in a Notch-independent manner.
Collapse
Affiliation(s)
- Tobias Friedrich
- Institute of Biochemistry, Justus-Liebig-University Giessen, Friedrichstrasse 24, 35392 Giessen, Germany.,Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany
| | - Francesca Ferrante
- Institute of Biochemistry, Justus-Liebig-University Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Léo Pioger
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS-UMR 5535, 1919 Route de Mende, 34293 cedex 5, Montpellier, France
| | - Andrea Nist
- Genomics Core Facility, Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Hans-Meerwein-Str. 3, 35043 Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Hans-Meerwein-Str. 3, 35043 Marburg, Germany
| | - Jean-Christophe Andrau
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS-UMR 5535, 1919 Route de Mende, 34293 cedex 5, Montpellier, France
| | - Marek Bartkuhn
- Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany.,Institute for Lung Health, Aulweg 132, 35392 Giessen, Germany
| | - Benedetto Daniele Giaimo
- Institute of Biochemistry, Justus-Liebig-University Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Tilman Borggrefe
- Institute of Biochemistry, Justus-Liebig-University Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| |
Collapse
|
6
|
Wu Y, Luo X, Zhou Q, Gong H, Gao H, Liu T, Chen J, Liang L, Kurihara H, Li YF, He RR. The disbalance of LRP1 and SIRP α by psychological stress dampens the clearance of tumor cells by macrophages. Acta Pharm Sin B 2022; 12:197-209. [PMID: 35127380 PMCID: PMC8799994 DOI: 10.1016/j.apsb.2021.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/11/2021] [Accepted: 05/19/2021] [Indexed: 12/22/2022] Open
Abstract
The relationship between chronic psychological stress and tumorigenesis has been well defined in epidemiological studies; however, the underlying mechanism remains underexplored. In this study, we discovered that impaired macrophage phagocytosis contributed to the psychological stress-evoked tumor susceptibility, and the stress hormone glucocorticoid (GC) was identified as a principal detrimental factor. Mechanistically, GC disturbed the balance of the “eat me” signal receptor (low-density lipoprotein receptor-related protein-1, LRP1) and the “don't eat me” signal receptor (signal regulatory protein alpha, SIRPα). Further analysis revealed that GC led to a direct, glucocorticoid receptor (GR)-dependent trans-repression of LRP1 expression, and the repressed LRP1, in turn, resulted in the elevated gene level of SIRPα by down-regulating miRNA-4695-3p. These data collectively demonstrate that stress induces the imbalance of the LRP1/SIRPα axis and entails the disturbance of tumor cell clearance by macrophages. Our findings provide the mechanistic insight into psychological stress-evoked tumor susceptibility and indicate that the balance of LRP1/SIRPα axis may serve as a potential therapeutic strategy for tumor treatment.
Collapse
Affiliation(s)
- Yanping Wu
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Xiang Luo
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Qingqing Zhou
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Haibiao Gong
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Huaying Gao
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Tongzheng Liu
- Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiaxu Chen
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
- Corresponding authors.
| | - Lei Liang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Hiroshi Kurihara
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yi-Fang Li
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Corresponding authors.
| | - Rong-Rong He
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
- Corresponding authors.
| |
Collapse
|
7
|
Wan B, Belghazi M, Lemauf S, Poirié M, Gatti JL. Proteomics of purified lamellocytes from Drosophila melanogaster HopT um-l identifies new membrane proteins and networks involved in their functions. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 134:103584. [PMID: 34033897 DOI: 10.1016/j.ibmb.2021.103584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 06/12/2023]
Abstract
In healthy Drosophila melanogaster larvae, plasmatocytes and crystal cells account for 95% and 5% of the hemocytes, respectively. A third type of hemocytes, lamellocytes, are rare, but their number increases after oviposition by parasitoid wasps. The lamellocytes form successive layers around the parasitoid egg, leading to its encapsulation and melanization, and finally the death of this intruder. However, the total number of lamellocytes per larva remains quite low even after parasitoid infestation, making direct biochemical studies difficult. Here, we used the HopTum-l mutant strain that constitutively produces large numbers of lamellocytes to set up a purification method and analyzed their major proteins by 2D gel electrophoresis and their plasma membrane surface proteins by 1D SDS-PAGE after affinity purification. Mass spectrometry identified 430 proteins from 2D spots and 344 affinity-purified proteins from 1D bands, for a total of 639 unique proteins. Known lamellocyte markers such as PPO3 and the myospheroid integrin were among the components identified with specific chaperone proteins. Affinity purification detected other integrins, as well as a wide range of integrin-associated proteins involved in the formation and function of cell-cell junctions. Overall, the newly identified proteins indicate that these cells are highly adapted to the encapsulation process (recognition, motility, adhesion, signaling), but may also have several other physiological functions (such as secretion and internalization of vesicles) under different signaling pathways. These results provide the basis for further in vivo and in vitro studies of lamellocytes, including the development of new markers to identify coexisting populations and their respective origins and functions in Drosophila immunity.
Collapse
Affiliation(s)
- Bin Wan
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Maya Belghazi
- Institute of NeuroPhysiopathology (INP), UMR7051, CNRS, Aix-Marseille Université, Marseille, 13015, France
| | - Séverine Lemauf
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Marylène Poirié
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Jean-Luc Gatti
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France.
| |
Collapse
|
8
|
Chen J, Su Y, Pi S, Hu B, Mao L. The Dual Role of Low-Density Lipoprotein Receptor-Related Protein 1 in Atherosclerosis. Front Cardiovasc Med 2021; 8:682389. [PMID: 34124208 PMCID: PMC8192809 DOI: 10.3389/fcvm.2021.682389] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
Low-density lipoprotein receptor–related protein-1 (LRP1) is a large endocytic and signaling receptor belonging to the LDL receptor (LDLR) gene family and that is widely expressed in several tissues. LRP1 comprises a large extracellular domain (ECD; 515 kDa, α chain) and a small intracellular domain (ICD; 85 kDa, β chain). The deletion of LRP1 leads to embryonic lethality in mice, revealing a crucial but yet undefined role in embryogenesis and development. LRP1 has been postulated to participate in numerous diverse physiological and pathological processes ranging from plasma lipoprotein homeostasis, atherosclerosis, tumor evolution, and fibrinolysis to neuronal regeneration and survival. Many studies using cultured cells and in vivo animal models have revealed the important roles of LRP1 in vascular remodeling, foam cell biology, inflammation and atherosclerosis. However, its role in atherosclerosis remains controversial. LRP1 not only participates in the removal of atherogenic lipoproteins and proatherogenic ligands in the liver but also mediates the uptake of aggregated LDL to promote the formation of macrophage- and vascular smooth muscle cell (VSMC)-derived foam cells, which causes a prothrombotic transformation of the vascular wall. The dual and opposing roles of LRP1 may also represent an interesting target for atherosclerosis therapeutics. This review highlights the influence of LRP1 during atherosclerosis development, focusing on its dual role in vascular cells and immune cells.
Collapse
Affiliation(s)
- Jiefang Chen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Su
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shulan Pi
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Nikolakopoulou AM, Wang Y, Ma Q, Sagare AP, Montagne A, Huuskonen MT, Rege SV, Kisler K, Dai Z, Körbelin J, Herz J, Zhao Z, Zlokovic BV. Endothelial LRP1 protects against neurodegeneration by blocking cyclophilin A. J Exp Med 2021; 218:211750. [PMID: 33533918 PMCID: PMC7863706 DOI: 10.1084/jem.20202207] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
The low-density lipoprotein receptor–related protein 1 (LRP1) is an endocytic and cell signaling transmembrane protein. Endothelial LRP1 clears proteinaceous toxins at the blood–brain barrier (BBB), regulates angiogenesis, and is increasingly reduced in Alzheimer’s disease associated with BBB breakdown and neurodegeneration. Whether loss of endothelial LRP1 plays a direct causative role in BBB breakdown and neurodegenerative changes remains elusive. Here, we show that LRP1 inactivation from the mouse endothelium results in progressive BBB breakdown, followed by neuron loss and cognitive deficits, which is reversible by endothelial-specific LRP1 gene therapy. LRP1 endothelial knockout led to a self-autonomous activation of the cyclophilin A–matrix metalloproteinase-9 pathway in the endothelium, causing loss of tight junctions underlying structural BBB impairment. Cyclophilin A inhibition in mice with endothelial-specific LRP1 knockout restored BBB integrity and reversed and prevented neuronal loss and behavioral deficits. Thus, endothelial LRP1 protects against neurodegeneration by inhibiting cyclophilin A, which has implications for the pathophysiology and treatment of neurodegeneration linked to vascular dysfunction.
Collapse
Affiliation(s)
- Angeliki Maria Nikolakopoulou
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Yaoming Wang
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Qingyi Ma
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Abhay P Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Axel Montagne
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Mikko T Huuskonen
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Sanket V Rege
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Zhonghua Dai
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Jakob Körbelin
- Hubertus Wald Cancer Center, Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joachim Herz
- Departments of Neuroscience, Molecular Genetics, and Neurology, University of Texas Southwestern Medical Center, Dallas, TX.,Center for Neuroscience, University of Freiburg, Freiburg, Germany
| | - Zhen Zhao
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA
| |
Collapse
|
10
|
Paunovska K, Da Silva Sanchez A, Foster MT, Loughrey D, Blanchard EL, Islam FZ, Gan Z, Mantalaris A, Santangelo PJ, Dahlman JE. Increased PIP3 activity blocks nanoparticle mRNA delivery. SCIENCE ADVANCES 2020; 6:eaba5672. [PMID: 32743074 PMCID: PMC7375820 DOI: 10.1126/sciadv.aba5672] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 06/05/2020] [Indexed: 05/06/2023]
Abstract
The biological pathways that affect drug delivery in vivo remain poorly understood. We hypothesized that altering cell metabolism with phosphatidylinositol (3,4,5)-triphosphate (PIP3), a bioactive lipid upstream of the metabolic pathway PI3K (phosphatidylinositol 3-kinase)/AKT/ mTOR (mammalian target of rapamycin) would transiently increase protein translated by nanoparticle-delivered messenger RNA (mRNA) since these pathways increase growth and proliferation. Instead, we found that PIP3 blocked delivery of clinically-relevant lipid nanoparticles (LNPs) across multiple cell types in vitro and in vivo. PIP3-driven reductions in LNP delivery were not caused by toxicity, cell uptake, or endosomal escape. Interestingly, RNA sequencing and metabolomics analyses suggested an increase in basal metabolic rate. Higher transcriptional activity and mitochondrial expansion led us to formulate two competing hypotheses that explain the reductions in LNP-mediated mRNA delivery. First, PIP3 induced consumption of limited cellular resources, "drowning out" exogenously-delivered mRNA. Second, PIP3 triggers a catabolic response that leads to protein degradation and decreased translation.
Collapse
Affiliation(s)
| | | | - Matthew T. Foster
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | | | - Fatima Z. Islam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zubao Gan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Athanasios Mantalaris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Philip J. Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | |
Collapse
|
11
|
Shin K, Cha Y, Ban YH, Seo DW, Choi EK, Park D, Kang SK, Ra JC, Kim YB. Anti-osteoarthritis effect of a combination treatment with human adipose tissue-derived mesenchymal stem cells and thrombospondin 2 in rabbits. World J Stem Cells 2019; 11:1115-1129. [PMID: 31875872 PMCID: PMC6904861 DOI: 10.4252/wjsc.v11.i12.1115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 10/14/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA), a chronic age-related disease characterized by the slowly progressive destruction of articular cartilage, is one of the leading causes of disability. As a new strategy for treatment of OA, mesenchymal stem cells (MSCs) have the potential for articular cartilage regeneration. Meanwhile, thrombospondin 2 (TSP2) promotes the chondrogenic differentiation of MSCs.
AIM To investigate whether TSP2 induces chondrogenic differentiation of human adipose-derived MSCs (hADMSCs) and potentiates the therapeutic effects of hADMSCs in OA rabbits.
METHODS We investigated the chondrogenic potential of TSP2 in hADMSCs by analyzing the expression of chondrogenic markers as well as NOTCH signaling genes in normal and TSP2 small interfering RNA (siRNA)-treated stem cells. Anterior cruciate ligament transection surgery was performed in male New Zealand white rabbits, and 8 wk later, hADMSCs (1.7 × 106 or 1.7 × 107 cells) were injected into the injured knees alone or in combination with intra-articular injection of TSP2 (100 ng/knee) at 2-d intervals. OA progression was monitored by gross, radiological, and histological examinations.
RESULTS In hADMSC culture, treatment with TSP2 increased the expression of chondrogenic markers (SOX9 and collagen II) as well as NOTCH signaling genes (JAGGED1 and NOTCH3), which were inhibited by TSP2 siRNA treatment. In vivo, OA rabbits treated with hADMSCs or TSP2 alone exhibited lower degree of cartilage degeneration, osteophyte formation, and extracellular matrix loss 8 wk after cell transplantation. Notably, such cartilage damage was further alleviated by the combination of hADMSCs and TSP2. In addition, synovial inflammatory cytokines, especially tumor-necrosis factor-α, markedly decreased following the combination treatment.
CONCLUSION The results indicate that TSP2 enhances chondrogenic differentiation of hADMSCs via JAGGED1/NOTCH3 signaling, and that combination therapy with hADMSCs and TSP2 exerts synergistic effects in the cartilage regeneration of OA joints.
Collapse
Affiliation(s)
- Kyungha Shin
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Yeseul Cha
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Young-Hwan Ban
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Da Woom Seo
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Ehn-Kyoung Choi
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Dongsun Park
- Department of Biology Education, Korea National University of Education, Cheongju 28173, Chungbuk, South Korea
| | - Sung Keun Kang
- Biostar Stem Cell Research Institute, R-BIO Co., Ltd., Seoul 07238, South Korea
| | - Jeong Chan Ra
- Biostar Stem Cell Research Institute, R-BIO Co., Ltd., Seoul 07238, South Korea
| | - Yun-Bae Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| |
Collapse
|
12
|
Pan W, Song XY, Hu QB, Zhang M, Xu XH. TSP2 acts as a suppresser of cell invasion, migration and angiogenesis in medulloblastoma by inhibiting the Notch signaling pathway. Brain Res 2019; 1718:223-230. [PMID: 31063715 DOI: 10.1016/j.brainres.2019.05.004] [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: 12/17/2018] [Revised: 04/10/2019] [Accepted: 05/04/2019] [Indexed: 10/26/2022]
Abstract
Medulloblastoma (MB) represents a fatal malignancy often occurring in children. Angiogenesis is a hallmark of the progression of MB. Over the past decade, investigators have attempted to develop more effective and less toxic anti-angiogenic strategies to treat MB. Thrombospondin (TSP) family is observed to be a key regulator of angiogenesis. Thus, the current study aimed to elucidate the function of TSP2 in patients with MB and the underlying mechanism. The expression of TSP2, Notch1 and VEGF in MB and adjacent tissues collected from clinical samples as well as a MB cell line (Daoy) was examined. The results demonstrated that in the MB tissues and Daoy cells, TSP2 was downregulated, while Notch1 and VEGF were upregulated. Then, after the Daoy cells were treated with TSP2 silencing, TSP2 overexpression, or Notch signaling pathway inhibition, a series of in vitro cell experiments were performed to verify the interaction between TSP2 and Notch signaling pathway, and to examine the abilities of cell proliferation, migration, invasion, and tube formation. Upregulation of TSP2 was observed to lead to the downregulation of the Notch signaling pathway. Moreover, cells overexpressing TSP2 exhibited diminished proliferation, invasion, migration, and tube formation. In addition, a significant attenuation of tumor growth and angiogenesis was identified in vivo in the Daoy cells overexpressing TSP2 inoculated in nude mice. Taken together, the key findings of this study revealed the inhibitory role of TSP2 in the development of MB via blockade of the Notch signaling pathway, highlighting its potential as a treatment target for MB treatment.
Collapse
Affiliation(s)
- Wei Pan
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Xing-Yu Song
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Qi-Bo Hu
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Meng Zhang
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Xiao-Heng Xu
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, PR China.
| |
Collapse
|
13
|
Chen W, Wu X, Yan X, Xu A, Yang A, You H. Multitranscriptome analyses reveal prioritized genes specifically associated with liver fibrosis progression independent of etiology. Am J Physiol Gastrointest Liver Physiol 2019; 316:G744-G754. [PMID: 30920297 DOI: 10.1152/ajpgi.00339.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Elimination or suppression of causative factors can raise the possibility of liver fibrosis regression. However, different injurious stimuli will give fibrosis from somewhat different etiologies, which, in turn, may hamper the discovery of liver fibrosis-specific therapeutic drugs. Therefore, the analogical cellular and molecular events shared by various etiology-evoked liver fibrosis should be clarified. Our present study systematically integrated five publicly available transcriptomic data sets regarding liver fibrosis with different etiologies from the Gene Expression Omnibus database and performed a series of bioinformatics analyses and experimental verifications. A total of 111 significantly upregulated and 16 downregulated genes were identified specific to liver fibrosis independent of any etiology. These genes were predominately enriched in some Kyoto Encyclopedia of Genes and Genomes pathways, including the "PI3K-AKT signaling pathway," "Focal adhesion," and "ECM-receptor interaction." Subsequently, five prioritized liver fibrosis-specific genes, including COL4A2, THBS2, ITGAV, LAMB1, and PDGFRA, were screened. These genes were positively associated with each other and liver fibrosis progression. In addition, they could robustly separate all stages of samples in both training and validation data sets with diverse etiologies when they were regarded as observed variables applied to principal component analysis plots. Expressions of all five genes were confirmed in activated primary mouse hepatic stellate cells (HSCs) and transforming growth factor β1-treated LX-2 cells. Moreover, THBS2 protein was enhanced in liver fibrosis rodent models, which could promote HSC activation and proliferation and facilitate NOTCH1/JAG1 expression in HSCs. Overall, our current study may provide potential targets for liver fibrosis therapy and aid to a deeper understanding of the molecular underpinnings of liver fibrosis. NEW & NOTEWORTHY Prioritized liver fibrosis-specific genes THBS2, COL4A2, ITGAV, LAMB1, and PDGFRA were identified and significantly associated with liver fibrosis progression and could be combined to discriminate liver fibrosis stages regardless of any etiology. Among the identified prioritized liver fibrosis-specific targets, THBS2 protein was confirmed to be enhanced in liver fibrosis rodent models, which could promote hepatic stellate cell (HSC) activation and proliferation and facilitate NOTCH1/JAG1 expression in HSCs.
Collapse
Affiliation(s)
- Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Xiaoning Wu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases , Beijing , China
| | - Xuzhen Yan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases , Beijing , China
| | - Anjian Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Aiting Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Hong You
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases , Beijing , China
| |
Collapse
|
14
|
Fuertes I, Jordão R, Piña B, Barata C. Time-dependent transcriptomic responses of Daphnia magna exposed to metabolic disruptors that enhanced storage lipid accumulation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 249:99-108. [PMID: 30884398 DOI: 10.1016/j.envpol.2019.02.102] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/31/2019] [Accepted: 02/28/2019] [Indexed: 05/03/2023]
Abstract
The analysis of lipid disruption in invertebrates is limited by our poor knowledge of their lipidomes and of the associated metabolic pathways. For example, the mechanism by which exposure of the crustacean Daphnia magna to tributyltin, juvenoids, or bisphenol A increase the accumulation of storage lipids into lipid droplets is largely unknown/presently unclear. Here we analyze transcriptome changes subsequent to this lipid accumulation effect induced by either the pesticide pyriproxyfen (a juvenoid agonist), the plasticizer bisphenol A, or the antifouling agent tributyltin. Changes in the whole transcriptome were assessed after 8 and 24 h of exposure, the period showing the greatest variation in storage lipid accumulation. The three compounds affected similarly to a total of 1388 genes (965 overexpressed and 423 underexpressed transcripts), but only after 24 h of exposure. In addition, 225 transcripts became up-regulated in samples exposed to tributyltin for both 8 h and 24 h. Using D. melanogaster functional annotation, we determined that upregulated genes were enriched in members of KEGG modules implicated in fatty acid, glycerophospholipid, and glycerolipid metabolic pathways, as well as in genes related to membrane constituents and to chitin and cuticle metabolic pathways. Conversely, down-regulated genes appeared mainly related to visual perception and to oocyte development signaling pathways. Many tributyltin specifically upregulated genes were related to neuro-active ligand receptor interaction signaling pathways. These changes were consistent with the phetotypic effects reported in this and in previous studies that exposure of D. magna to the tested compounds increased lipid accumulation and reduced egg quantity and quality.
Collapse
Affiliation(s)
- Inmaculada Fuertes
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA), Spanish Research Council (IDAEA, CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Rita Jordão
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA), Spanish Research Council (IDAEA, CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Benjamín Piña
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA), Spanish Research Council (IDAEA, CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Carlos Barata
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA), Spanish Research Council (IDAEA, CSIC), Jordi Girona 18, 08034, Barcelona, Spain.
| |
Collapse
|
15
|
Bres EE, Faissner A. Low Density Receptor-Related Protein 1 Interactions With the Extracellular Matrix: More Than Meets the Eye. Front Cell Dev Biol 2019; 7:31. [PMID: 30931303 PMCID: PMC6428713 DOI: 10.3389/fcell.2019.00031] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a biological substrate composed of collagens, proteoglycans and glycoproteins that ensures proper cell migration and adhesion and keeps the cell architecture intact. The regulation of the ECM composition is a vital process strictly controlled by, among others, proteases, growth factors and adhesion receptors. As it appears, ECM remodeling is also essential for proper neuronal and glial development and the establishment of adequate synaptic signaling. Hence, disturbances in ECM functioning are often present in neurodegenerative diseases like Alzheimer’s disease. Moreover, mutations in ECM molecules are found in some forms of epilepsy and malfunctioning of ECM-related genes and pathways can be seen in, for example, cancer or ischemic injury. Low density lipoprotein receptor-related protein 1 (Lrp1) is a member of the low density lipoprotein receptor family. Lrp1 is involved not only in ligand uptake, receptor mediated endocytosis and lipoprotein transport—functions shared by low density lipoprotein receptor family members—but also regulates cell surface protease activity, controls cellular entry and binding of toxins and viruses, protects against atherosclerosis and acts on many cell signaling pathways. Given the plethora of functions, it is not surprising that Lrp1 also impacts the ECM and is involved in its remodeling. This review focuses on the role of Lrp1 and some of its major ligands on ECM function. Specifically, interactions with two Lrp1 ligands, integrins and tissue plasminogen activator are described in more detail.
Collapse
Affiliation(s)
- Ewa E Bres
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
16
|
Lommel M, Strompen J, Hellewell AL, Balasubramanian GP, Christofidou ED, Thomson AR, Boyle AL, Woolfson DN, Puglisi K, Hartl M, Holstein TW, Adams JC, Özbek S. Hydra Mesoglea Proteome Identifies Thrombospondin as a Conserved Component Active in Head Organizer Restriction. Sci Rep 2018; 8:11753. [PMID: 30082916 PMCID: PMC6079037 DOI: 10.1038/s41598-018-30035-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 07/23/2018] [Indexed: 02/06/2023] Open
Abstract
Thrombospondins (TSPs) are multidomain glycoproteins with complex matricellular functions in tissue homeostasis and remodeling. We describe a novel role of TSP as a Wnt signaling target in the basal eumetazoan Hydra. Proteome analysis identified Hydra magnipapillata TSP (HmTSP) as a major component of the cnidarian mesoglea. In general, the domain organization of cnidarian TSPs is related to the pentameric TSPs of bilaterians, and in phylogenetic analyses cnidarian TSPs formed a separate clade of high sequence diversity. HmTSP expression in polyps was restricted to the hypostomal tip and tentacle bases that harbor Wnt-regulated organizer tissues. In the hypostome, HmTSP- and Wnt3-expressing cells were identical or in close vicinity to each other, and regions of ectopic tentacle formation induced by pharmacological β-Catenin activation (Alsterpaullone) corresponded to foci of HmTSP expression. Chromatin immunoprecipitation (ChIP) confirmed binding of Hydra TCF to conserved elements in the HmTSP promotor region. Accordingly, β-Catenin knockdown by siRNAs reduced normal HmTSP expression at the head organizer. In contrast, knockdown of HmTSP expression led to increased numbers of ectopic organizers in Alsterpaullone-treated animals, indicating a negative regulatory function. Our data suggest an unexpected role for HmTSP as a feedback inhibitor of Wnt signaling during Hydra body axis patterning and maintenance.
Collapse
Affiliation(s)
- Mark Lommel
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Jennifer Strompen
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Andrew L Hellewell
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Gnana Prakash Balasubramanian
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany.,G200 Division of Applied Bioinformatics, German Cancer Research Institute (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany
| | - Elena D Christofidou
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Andrew R Thomson
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, BS8 1TS, UK.,School of Chemistry, University of Glasgow, Joseph Black Building, University Avenue, Glasgow, G12 8QQ, Scotland
| | - Aimee L Boyle
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.,School of Chemistry, Cantock's Close, University of Bristol, Bristol, BS8 1TS, UK.,Leiden Institute of Chemistry, Leiden University, POB 9502, NL-2300, RA Leiden, Netherlands
| | - Derek N Woolfson
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.,School of Chemistry, Cantock's Close, University of Bristol, Bristol, BS8 1TS, UK
| | - Kane Puglisi
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria
| | - Markus Hartl
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria
| | - Thomas W Holstein
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Josephine C Adams
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| | - Suat Özbek
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany.
| |
Collapse
|
17
|
Lactoferrin-induced growth factors and cytokines expression profile in pre-osteoblast MC3T3-E1 cell and LRP1 stable knockdown MC3T3-E1 cell. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
18
|
Jeong SY, Ha J, Lee M, Jin HJ, Kim DH, Choi SJ, Oh W, Yang YS, Kim JS, Kim BG, Chang JH, Cho DH, Jeon HB. Autocrine Action of Thrombospondin-2 Determines the Chondrogenic Differentiation Potential and Suppresses Hypertrophic Maturation of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells. Stem Cells 2015; 33:3291-303. [PMID: 26235673 DOI: 10.1002/stem.2120] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 06/30/2015] [Indexed: 12/13/2022]
Abstract
Previous studies have shown that mesenchymal stem cell (MSC)-based therapies have varying efficacies for the treatment of various diseases, including cartilage defects. In this study, we demonstrated that the chondrogenic differentiation potential of human umbilical cord blood-derived MSCs (hUCB-MSCs) obtained from different individual donors varies, and we investigated the molecular basis for this variation. Microarray gene expression analysis identified thrombospondin-2 (TSP2) as a candidate gene underlying the interindividual variation in the chondrogenic differentiation potential of hUCB-MSCs. To assess the association between TSP-2 and the differentiation potential, we evaluated chondrogenic differentiation of hUCB-MSCs treated with TSP2 siRNA. In addition, we studied the effect of supplementing exogenous recombinant TSP-2 on TSP2 siRNA-treated hUCB-MSCs. We found that TSP-2 autocrinally promoted chondrogenic differentiation of hUCB-MSCs via the Notch signaling pathway, which was confirmed in MSCs from other sources such as bone marrow and adipose tissue. Interestingly, we observed that TSP-2 attenuated hypertrophy, which inevitably occurs during chondrogenic differentiation of hUCB-MSCs. Our findings indicated that the variable chondrogenic differentiation potential of MSCs obtained from different donors is influenced by the TSP-2 level in the differentiating cells. Thus, the TSP-2 level can be used as a marker to select MSCs with superior chondrogenic differentiation potential for use in cartilage regeneration therapy.
Collapse
Affiliation(s)
- Sang Young Jeong
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Jueun Ha
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Miyoung Lee
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Hye Jin Jin
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Dong Hyun Kim
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Soo Jin Choi
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Wonil Oh
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Jae-Sung Kim
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Byung-Gyu Kim
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, School of Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jeong Ho Chang
- Department of Biology, Teachers College, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Hyung Cho
- Graduate School of East-West Medical Science, Kyung Hee University, Seoul, Republic of Korea
| | - Hong Bae Jeon
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| |
Collapse
|
19
|
Bioactive nanofibers enable the identification of thrombospondin 2 as a key player in enamel regeneration. Biomaterials 2015; 61:216-28. [PMID: 26004236 DOI: 10.1016/j.biomaterials.2015.05.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/14/2015] [Accepted: 05/18/2015] [Indexed: 12/19/2022]
Abstract
Tissue regeneration and development involves highly synchronized signals both between cells and with the extracellular environment. Biomaterials can be tuned to mimic specific biological signals and control cell response(s). As a result, these materials can be used as tools to elucidate cell signaling pathways and candidate molecules involved with cellular processes. In this work, we explore enamel-forming cells, ameloblasts, which have a limited regenerative capacity. By exposing undifferentiated cells to a self-assembling matrix bearing RGDS epitopes, we elicited a regenerative signal at will that subsequently led to the identification of thrombospondin 2 (TSP2), an extracellular matrix protein that has not been previously recognized as a key player in enamel development and regeneration. Targeted disruption of the thrombospondin 2 gene (Thbs2) resulted in enamel formation with a disordered architecture that was highly susceptible to wear compared to their wild-type counterparts. To test the regenerative capacity, we injected the bioactive matrix into the enamel organ and discovered that the enamel organic epithelial cells in TSP-null mice failed to polarize on the surface of the artificial matrix, greatly reducing integrin β1 and Notch1 expression levels, which represent signaling pathways known to be associated with TSP2. These results suggest TSP2 plays an important role in regulating cell-matrix interactions during enamel formation. Exploiting the signaling pathways activated by biomaterials can provide insight into native signaling mechanisms crucial for tooth development and cell-based strategies for enamel regeneration.
Collapse
|
20
|
Wang B, Guo W, Huang Y. Thrombospondins and synaptogenesis. Neural Regen Res 2015; 7:1737-43. [PMID: 25624796 PMCID: PMC4302456 DOI: 10.3969/j.issn.1673-5374.2012.22.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/03/2012] [Indexed: 12/28/2022] Open
Abstract
Here, we review research on the mechanisms underlying the ability of thrombospondin to promote synaptogenesis and examine its role in central nervous system diseases and drug actions. Thrombospondin secreted by glial cells plays a critical role in synaptogenesis and maintains synapse stability. Thrombospondin regulates synaptogenesis through receptor α2δ-1 and neuroligin 1, and promotes the proliferation and differentiation of neural progenitor cells. It also participates in synaptic remodeling following injury and in the action of some nervous system drugs.
Collapse
Affiliation(s)
- Bin Wang
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, Guangdong Province, China
| | - Weitao Guo
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, Guangdong Province, China
| | - Yun Huang
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, Guangdong Province, China
| |
Collapse
|
21
|
The small leucine-rich proteoglycan BGN accumulates in CADASIL and binds to NOTCH3. Transl Stroke Res 2015; 6:148-55. [PMID: 25578324 DOI: 10.1007/s12975-014-0379-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/09/2014] [Accepted: 11/18/2014] [Indexed: 10/24/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an inherited form of cerebral small vessel disease caused by mutations in conserved residues of NOTCH3. Affected arteries of CADASIL feature fibrosis and accumulation of NOTCH3. A variety of collagen subtypes (types I, III, IV, and VI) have been identified in fibrotic CADASIL vessels. Biglycan (BGN) and decorin (DCN) are class I members of the small leucine-rich proteoglycan (SLRP) family that regulate collagen fibril size. Because DCN has been shown to deposit in arteries in cerebral small vessel disease, we tested whether BGN accumulates in arteries of CADASIL brains. BGN was strongly expressed in both small penetrating and leptomeningeal arteries of CADASIL brain. BGN protein was localized to all three layers of arteries (intima, media, and adventitia). Substantially, more immunoreactivity was observed in CADASIL brains compared to controls. Immunoblotting of brain lysates showed a fourfold increase in CADASIL brains (compared to controls). Messenger RNA encoding BGN was also increased in CADASIL and was localized by in situ hybridization to all three vascular layers in CADASIL. Human cerebrovascular smooth muscle cells exposed to purified NOTCH3 ectodomain upregulated BGN, DCN, and COL4A1 through mechanisms that are sensitive to rapamycin, a potent mTOR inhibitor. In addition, BGN protein interacted directly with NOTCH3 protein in cell culture and in direct protein interaction assays. In conclusion, BGN is a CADASIL-enriched protein that potentially accumulates in vessels by mTOR-mediated transcriptional activation and/or post-translational accumulation via protein interactions with NOTCH3 and collagen.
Collapse
|
22
|
Zhang X, Lee SJ, Young KZ, Josephson DA, Geschwind MD, Wang MM. Latent NOTCH3 epitopes unmasked in CADASIL and regulated by protein redox state. Brain Res 2014; 1583:230-6. [PMID: 25150590 PMCID: PMC4206828 DOI: 10.1016/j.brainres.2014.08.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 08/07/2014] [Indexed: 12/29/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy CADASIL is caused by more than a hundred NOTCH3 mutations. Virtually all encoded mutant proteins contain an odd number of cysteines. As such, structural changes in NOTCH3 may be the primary molecular abnormality in CADASIL. Thus, we sought evidence for structurally altered NOTCH3 protein in CADASIL tissue. Four antibodies were raised in rabbits against two non-overlapping N-terminal NOTCH3 sequences. These reagents were used in immunohistochemical experiments to detect epitopes in post-mortem CADASIL brains (n=8), control brains, and cells overexpressing NOTCH3. To determine the biochemical nature of NOTCH3 epitopes, we used these antibodies to probe pure NOTCH3-Fc fusion proteins treated with acid, urea, guanidinium, ionic detergents, acrylamide, and thiol- and phosphorus-based reductants. All antibodies avidly stained arteries in 8 of 8 CADASIL brain samples. The most prominent staining was in degenerating media of leptomeningeal arteries and sclerotic penetrating vessels. Normal appearing vessels from control brains were not reactive. Antibodies did not react with cultured cells overexpressing NOTCH3 or with purified NOTCH3-Fc protein. Furthermore, treatment of pure protein with acid, chaotropic denaturants, alkylators, and detergents failed to unmask N-terminal NOTCH3 epitopes. Antibodies, however, recognized novel N-terminal epitopes in purified NOTCH3-Fc protein treated with three different reductants (DTT, beta-mercaptoethanol, and TCEP). We conclude that CADASIL arteries feature latent N-terminal NOTCH3 epitopes, suggesting the first evidence in vivo of NOTCH3 structural alterations.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Departments of Neurology, University of Michigan, Ann Arbor, MI 48109-5622, USA
| | - Soo Jung Lee
- Departments of Neurology, University of Michigan, Ann Arbor, MI 48109-5622, USA
| | - Kelly Z Young
- Departments of Neurology, University of Michigan, Ann Arbor, MI 48109-5622, USA
| | - David A Josephson
- Josephson-Wallack-Munshower Neurology, Indianapolis, IN 46260, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46260, USA
| | - Michael D Geschwind
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA 94143, USA
| | - Michael M Wang
- Departments of Neurology, University of Michigan, Ann Arbor, MI 48109-5622, USA; Molecular and Integrative Physiology, University of Michigan, 7725 Med Sci II 5622, 1137 Catherine Street, Ann Arbor, MI 48109-5622, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA.
| |
Collapse
|
23
|
Abstract
Small penetrating brain artery thickening is a major feature of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Although affected fibrotic arteries of CADASIL have been shown to accumulate collagen, other components that compose pathological arterial walls remain incompletely characterized. We investigated the expression of decorin (DCN), the first collagen-binding small leucine-rich proteoglycan identified, in CADASIL. DCN was markedly upregulated in pathologically affected leptomeningeal and small penetrating arteries in CADASIL and was notably weaker in normal arteries from control brains. DCN protein was localized principally to the media and adventitia and only occasionally expressed in the intima. Immunoblotting of brain lysates showed a three-fold increase of DCN in CADASIL brains (compared with controls). Messenger RNA encoding DCN was five-fold increased in CADASIL. We conclude that DCN is the first identified proteoglycan to be identified in CADASIL arteries and may accumulate through transcriptional mechanisms. Additional studies are warranted to determine whether DCN localizes broadly to pathological small vessels in other cerebrovascular disorders.
Collapse
Affiliation(s)
- Soo Jung Lee
- Departments of aNeurology bMolecular & Integrative Physiology, University of Michigan cNeurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
24
|
Phamduong E, Rathore MK, Crews NR, D’Angelo AS, Leinweber AL, Kappera P, Krenning TM, Rendell VR, Belcheva MM, Coscia CJ. Acute and chronic mu opioids differentially regulate thrombospondins 1 and 2 isoforms in astrocytes. ACS Chem Neurosci 2014; 5:106-14. [PMID: 24304333 DOI: 10.1021/cn400172n] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Chronic opioids induce synaptic plasticity, a major neuronal adaptation. Astrocyte activation in synaptogenesis may play a critical role in opioid tolerance, withdrawal, and dependence. Thrombospondins 1 and 2 (TSP1/2) are astrocyte-secreted matricellular glycoproteins that promote neurite outgrowth as well as dendritic spine and synapse formation, all of which are inhibited by chronic μ opioids. In prior studies, we discovered that the mechanism of TSP1 regulation by μ opioids in astrocytes involves crosstalk between three different classes of receptors, μ opioid receptor, EGFR and TGFβR. Moreover, TGFβ1 stimulated TSP1 expression via EGFR and ERK/MAPK activation, indicating that EGFR is a signaling hub for opioid and TGFβ1 actions. Using various selective antagonists, and inhibitors, here we compared the mechanisms of chronic opioid regulation of TSP1/2 isoform expression in vivo and in immortalized rat cortical astrocytes. TSP1/2 release from astrocytes was also monitored. Acute and chronic μ opioids, morphine, and the prototypic μ ligand, DAMGO, modulated TSP2 protein levels. TSP2 but not TSP1 protein content was up-regulated by acute (3 h) morphine or DAMGO by an ERK/MAPK dependent mechanism. Paradoxically, TSP2 protein levels were altered neither by TGFβ1 nor by astrocytic neurotrophic factors, EGF, CNTF, and BMP4. TSP1/2 immunofluorescence was increased in astrocytes subjected to scratch-wounding, suggesting TSPs may be useful markers for the "reactive" state of these cells and potentially for different types of injury. Previously, we determined that chronic morphine attenuated both neurite outgrowth and synapse formation in cocultures of primary astrocytes and neurons under similar temporal conditions that μ opioids reduced TSP1 protein levels in astrocytes. Here we found that, after the same 8 day treatment, morphine or DAMGO diminished TSP2 protein levels in astrocytes. Therefore, μ opioids may deter synaptogenesis via both TSP1/2 isoforms, but by distinct mechanisms.
Collapse
Affiliation(s)
- Ellen Phamduong
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Maanjot K. Rathore
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Nicholas R. Crews
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Alexander S. D’Angelo
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Andrew L. Leinweber
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Pranay Kappera
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Thomas M. Krenning
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Victoria R. Rendell
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Mariana M. Belcheva
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Carmine J. Coscia
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| |
Collapse
|
25
|
Meng H, Zhang X, Lee SJ, Wang MM. Von Willebrand factor inhibits mature smooth muscle gene expression through impairment of Notch signaling. PLoS One 2013; 8:e75808. [PMID: 24086636 PMCID: PMC3781053 DOI: 10.1371/journal.pone.0075808] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 08/20/2013] [Indexed: 12/30/2022] Open
Abstract
Von Willebrand factor (vWF), a hemostatic protein normally synthesized and stored by endothelial cells and platelets, has been localized beyond the endothelium in vascular disease states. Previous studies have implicated potential non-hemostatic functions of vWF, but signaling mechanisms underlying its effects are currently undefined. We present evidence that vWF breaches the endothelium and is expressed in a transmural distribution pattern in cerebral small vessel disease (SVD). To determine the potential molecular consequences of vWF permeation into the vessel wall, we also tested whether vWF impairs Notch regulation of key smooth muscle marker genes. In a co-culture system using Notch ligand expressing cells to stimulate Notch in A7R5 cells, vWF strongly inhibited both the Notch pathway and the activation of mature smooth muscle gene promoters. Similar repressive effects were observed in primary human cerebral vascular smooth muscle cells. Expression of the intracellular domain of NOTCH3 allowed cells to bypass the inhibitory effects of vWF. Moreover, vWF forms molecular complexes with all four mammalian Notch ectodomains, suggesting a novel function of vWF as an extracellular inhibitor of Notch signaling. In sum, these studies demonstrate vWF in the vessel wall as a common feature of cerebral SVD; furthermore, we provide a plausible mechanism by which non-hemostatic vWF may play a novel role in the promotion of vascular disease.
Collapse
Affiliation(s)
- He Meng
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Xiaojie Zhang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Soo Jung Lee
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Michael M. Wang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
26
|
Zhang X, Meng H, Wang MM. Collagen represses canonical Notch signaling and binds to Notch ectodomain. Int J Biochem Cell Biol 2013; 45:1274-80. [PMID: 23579095 PMCID: PMC3683383 DOI: 10.1016/j.biocel.2013.03.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 03/16/2013] [Accepted: 03/29/2013] [Indexed: 12/27/2022]
Abstract
The Notch signaling system features a growing number of modulators that include extracellular proteins that bind to the Notch ectodomain. Collagens are a complex, heterogeneous family of secreted proteins that serve both structural and signaling functions, most prominently through binding to integrins and DDR. The shared widespread tissue distribution of Notch and collagen prompted us to investigate the effects of collagen on Notch signaling. In a cell co-culture signaling assay, we found that type IV collagen inhibited Notch signaling in H460 and A7R5 cell lines. Moreover, Notch-stimulated expression of mature smooth muscle genes SMA, MHC, SM22, and calponin, which define the physiologic phenotype of normal vascular smooth muscle, was inhibited by type IV collagen in A7R5 cells. Cloned promoters of three of these genes were also inhibited by exposure to collagen. Collagen-dependent repression of Notch signaling required an RBP-jK site within the SM22 promoter. Moreover, repression by collagen required extracellular stimulation of the Notch signaling pathway. Type IV collagen bound to both Notch3 and Jagged1 proteins in purified protein binding assays. In addition, type I collagen also inhibited Notch signaling and bound to Notch and Jagged. We conclude that type IV and type I collagen repress canonical Notch signaling to alter expression of Notch target genes.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-5622, USA
| | | | | |
Collapse
|
27
|
Hennen E, Safina D, Haussmann U, Wörsdörfer P, Edenhofer F, Poetsch A, Faissner A. A LewisX glycoprotein screen identifies the low density lipoprotein receptor-related protein 1 (LRP1) as a modulator of oligodendrogenesis in mice. J Biol Chem 2013; 288:16538-16545. [PMID: 23615909 DOI: 10.1074/jbc.m112.419812] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In the developing and adult CNS multipotent neural stem cells reside in distinct niches. Specific carbohydrates and glycoproteins are expressed in these niche microenvironments which are important regulators of stem cell maintenance and differentiation fate. LewisX (LeX), also known as stage-specific embryonic antigen-1 or CD15, is a defined carbohydrate moiety expressed in niche microenvironments of the developing and adult CNS. LeX-glycans are involved in stem cell proliferation, migration, and stemness. A few LeX carrier proteins are known, but a systematic analysis of the targets of LeX glycosylation in vivo has not been performed so far. Using LeX glycosylation as a biomarker we aimed to discover new glycoproteins with a potential functional relevance for CNS development. By immunoaffinity chromatography we enriched LeX glycoproteins from embryonic and postnatal mouse brains and used one-dimensional nLC-ESI-MS/MS for their identification. We could validate phosphacan, tenascin-C, and L1-CAM as major LeX carrier proteins present in vivo. Furthermore, we identified LRP1, a member of the LDL receptor family, as a new LeX carrier protein expressed by mouse neural stem cells. Surprisingly, little is known about LRP1 function for neural stem cells. Thus, we generated Lrp1 knock-out neural stem cells by Cre-mediated recombination and investigated their properties. Here, we provide first evidence that LRP1 is necessary for the differentiation of neural stem cells toward oligodendrocytes. However, this function is independent of LeX glycosylation.
Collapse
Affiliation(s)
- Eva Hennen
- Departments of Cell Morphology and Molecular Neurobiology, D-44780 Bochum, Germany
| | - Dina Safina
- Departments of Cell Morphology and Molecular Neurobiology, D-44780 Bochum, Germany
| | - Ute Haussmann
- Plant Biochemistry, Ruhr-University Bochum, D-44780 Bochum, Germany
| | - Philipp Wörsdörfer
- Stem Cell Engineering Group, Institute of Reconstructive Neurobiology, University of Bonn-Life and Brain Center, D-53105 Bonn, Germany
| | - Frank Edenhofer
- Stem Cell Engineering Group, Institute of Reconstructive Neurobiology, University of Bonn-Life and Brain Center, D-53105 Bonn, Germany
| | - Ansgar Poetsch
- Plant Biochemistry, Ruhr-University Bochum, D-44780 Bochum, Germany
| | - Andreas Faissner
- Departments of Cell Morphology and Molecular Neurobiology, D-44780 Bochum, Germany.
| |
Collapse
|
28
|
Meng H, Zhang X, Yu G, Lee SJ, Chen YE, Prudovsky I, Wang MM. Biochemical characterization and cellular effects of CADASIL mutants of NOTCH3. PLoS One 2012; 7:e44964. [PMID: 23028706 PMCID: PMC3445613 DOI: 10.1371/journal.pone.0044964] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 08/16/2012] [Indexed: 12/16/2022] Open
Abstract
Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is the best understood cause of dominantly inherited stroke and results from NOTCH3 mutations that lead to NOTCH3 protein accumulation and selective arterial smooth muscle degeneration. Previous studies show that NOTCH3 protein forms multimers. Here, we investigate protein interactions between NOTCH3 and other vascular Notch isoforms and characterize the effects of elevated NOTCH3 on smooth muscle gene regulation. We demonstrate that NOTCH3 forms heterodimers with NOTCH1, NOTCH3, and NOTCH4. R90C and C49Y mutant NOTCH3 form complexes which are more resistant to detergents than wild type NOTCH3 complexes. Using quantitative NOTCH3-luciferase clearance assays, we found significant inhibition of mutant NOTCH3 clearance. In coculture assays of NOTCH function, overexpressed wild type and mutant NOTCH3 significantly repressed NOTCH-regulated smooth muscle transcripts and potently impaired the activity of three independent smooth muscle promoters. Wildtype and R90C recombinant NOTCH3 proteins applied to cell cultures also blocked canonical Notch fuction. We conclude that CADASIL mutants of NOTCH3 complex with NOTCH1, 3, and 4, slow NOTCH3 clearance, and that overexpressed wild type and mutant NOTCH3 protein interfere with key NOTCH-mediated functions in smooth muscle cells.
Collapse
Affiliation(s)
- He Meng
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Xiaojie Zhang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Genggeng Yu
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Soo Jung Lee
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Y. Eugene Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Igor Prudovsky
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, United States of America
| | - Michael M. Wang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
29
|
Van Hul M, Frederix L, Lijnen HR. Role of thrombospondin-2 in murine adipose tissue angiogenesis and development. Obesity (Silver Spring) 2012; 20:1757-62. [PMID: 21818150 DOI: 10.1038/oby.2011.260] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Expression of thrombospondin-2 (TSP-2), a matricellular protein with anti-angiogenic properties, is modulated in developing adipose tissue. To investigate a potential functional role of TSP-2 in adipose tissue angiogenesis and growth, TSP-2 deficient (TSP-2(-/-)) and wild-type littermate (TSP-2(+/+)) mice were kept on normal chow (standard fat diet (SFD)) or on high fat diet (HFD) for 15 weeks. TSP-2(-/-) mice kept on HFD had a significantly lower total body weight throughout the experimental period. Subcutaneous (SC) and gonadal (GON) fat mass were, however, not different, and their composition in terms of size and density of adipocytes and blood vessels was also comparable in both genotypes. Macrophage infiltration in SC or GON adipose tissues was not affected by TSP-2 deficiency. TSP-2 deficiency had no effect on adipose tissue mRNA expression of gelatinase A (MMP-2), whereas gelatinase B (MMP-9) was downregulated in SC and GON adipose tissues of TSP-2(-/-) mice on HFD. Glucose tolerance and insulin resistance tests were comparable for TSP-2(+/+) and TSP-2(-/-) mice. TSP-2 deficiency was not compensated by increased expression of TSP-1 in the TSP-2(-/-) mice. These data suggest that TSP-2, despite its reported anti-angiogenic properties, does not play an important functional role in adipose tissue related angiogenesis or associated fat development in mice.
Collapse
Affiliation(s)
- Matthias Van Hul
- Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
30
|
Risher WC, Eroglu C. Thrombospondins as key regulators of synaptogenesis in the central nervous system. Matrix Biol 2012; 31:170-7. [PMID: 22285841 DOI: 10.1016/j.matbio.2012.01.004] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 01/04/2012] [Accepted: 01/04/2012] [Indexed: 01/07/2023]
Abstract
Thrombospondins (TSPs) are a family of large, oligomeric multidomain glycoproteins that participate in a variety of biological functions as part of the extracellular matrix (ECM). Through their associations with a number of binding partners, TSPs mediate complex cell-cell and cell-matrix interactions in such diverse processes as angiogenesis, inflammation, osteogenesis, cell proliferation, and apoptosis. It was recently shown in the developing central nervous system (CNS) that TSPs promote the formation of new synapses, which are the unique cell-cell adhesions between neurons in the brain. This increase in synaptogenesis is mediated by the interaction between astrocyte-secreted TSPs and their neuronal receptor, calcium channel subunit α2δ-1. The cellular and molecular mechanisms that underlie induction of synaptogenesis via this interaction are yet to be fully elucidated. This review will focus on what is known about TSP and synapse formation during development, possible roles for TSP following brain injury, and what the previously established actions of TSP in other biological tissues may tell us about the mechanisms underlying TSP's functions in CNS synaptogenesis.
Collapse
Affiliation(s)
- W Christopher Risher
- Cell Biology Department, Duke University Medical Center, Durham, NC 27710, United States
| | | |
Collapse
|
31
|
Von Willebrand Factor permeates small vessels in CADASIL and inhibits smooth muscle gene expression. Transl Stroke Res 2011; 3:138-45. [PMID: 22639698 DOI: 10.1007/s12975-011-0112-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
BACKGROUND AND PURPOSE CADASIL (cerebral autosomal dominant arteriopathy subcortical infarcts and leukoencephalopathy) is a genetic disorder hallmarked by ischemic stroke and vascular dementia. Characteristic pathological changes in the vasculature include thickening of small arteries and accumulation of heterogeneous material within the vessel wall. We tested whether endothelial von Willebrand factor (vWF) accumulates in CADASIL vessels and whether exposure of smooth muscle cells to vWF alters the expression of smooth muscle gene expression. METHODS Brain sections obtained at autopsy from six North American CADASIL patients were examined using immunohistochemistry for vWF and IgG. Rat aortic smooth muscle cells (A7R5 cells) were tested for binding to infrared-tag labeled vWF. Finally, A7R5 cells were exposed to vWF, and expression of mature smooth muscle marker genes was analyzed by quantitative reverse transcriptase PCR. RESULTS vWF is expressed in the penetrating arterial walls in all CADASIL samples. IgG, a marker of serum extravasation, was present only in a minority of arterial walls. vWF binds to smooth muscle cells in vitro, and low concentrations of vWF rapidly activate c-fos, EGR, TSP1, and c-myc while specifically inhibiting RNA encoding smooth muscle actin, calponin, and SM22. CONCLUSIONS These data demonstrate that vWF, likely produced by the endothelium, permeates the vessel wall of CADASIL brains. Exposure of smooth muscle cells to vWF results in reduction of specific RNAs required for normal vascular homeostasis. This is the first report of accumulation of a protein within CADASIL vessels that inhibits vascular gene expression and implicates a role for vWF beyond hemostasis.
Collapse
|
32
|
Abstract
Thrombospondins are evolutionarily conserved, calcium-binding glycoproteins that undergo transient or longer-term interactions with other extracellular matrix components. They share properties with other matrix molecules, cytokines, adaptor proteins, and chaperones, modulate the organization of collagen fibrils, and bind and localize an array of growth factors or proteases. At cell surfaces, interactions with an array of receptors activate cell-dependent signaling and phenotypic outcomes. Through these dynamic, pleiotropic, and context-dependent pathways, mammalian thrombospondins contribute to wound healing and angiogenesis, vessel wall biology, connective tissue organization, and synaptogenesis. We overview the domain organization and structure of thrombospondins, key features of their evolution, and their cell biology. We discuss their roles in vivo, associations with human disease, and ongoing translational applications. In many respects, we are only beginning to appreciate the important roles of these proteins in physiology and pathology.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom.
| | | |
Collapse
|
33
|
Folkersen L, Wågsäter D, Paloschi V, Jackson V, Petrini J, Kurtovic S, Maleki S, Eriksson MJ, Caidahl K, Hamsten A, Michel JB, Liska J, Gabrielsen A, Franco-Cereceda A, Eriksson P. Unraveling divergent gene expression profiles in bicuspid and tricuspid aortic valve patients with thoracic aortic dilatation: the ASAP study. Mol Med 2011; 17:1365-73. [PMID: 21968790 DOI: 10.2119/molmed.2011.00286] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 09/20/2011] [Indexed: 01/12/2023] Open
Abstract
Thoracic aortic aneurysm (TAA) is a common complication in patients with a bicuspid aortic valve (BAV), the most frequent congenital heart disorder. For unknown reasons TAA occurs at a younger age, with a higher frequency in BAV patients than in patients with a tricuspid aortic valve (TAV), resulting in an increased risk for aortic dissection and rupture. To investigate the increased TAA incidence in BAV patients, we obtained tissue biopsy samples from nondilated and dilated aortas of 131 BAV and TAV patients. Global gene expression profiles were analyzed from controls and from aortic intima-media and adventitia of patients (in total 345 samples). Of the genes found to be differentially expressed with dilation, only a few (<4%) were differentially expressed in both BAV and TAV patients. With the use of gene set enrichment analysis, the cell adhesion and extracellular region gene ontology sets were identified as common features of TAA in both BAV and TAV patients. Immune response genes were observed to be particularly overexpressed in the aortic media of dilated TAV samples. The divergent gene expression profiles indicate that there are fundamental differences in TAA etiology in BAV and TAV patients. Immune response activation solely in the aortic media of TAV patients suggests that inflammation is involved in TAA formation in TAV but not in BAV patients. Conversely, genes were identified that were only differentially expressed with dilation in BAV patients. The result has bearing on future clinical studies in which separate analysis of BAV and TAV patients is recommended.
Collapse
Affiliation(s)
- Lasse Folkersen
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wang MM. Notch signaling and Notch signaling modifiers. Int J Biochem Cell Biol 2011; 43:1550-62. [PMID: 21854867 DOI: 10.1016/j.biocel.2011.08.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 07/28/2011] [Accepted: 08/05/2011] [Indexed: 02/07/2023]
Abstract
Originally discovered nearly a century ago, the Notch signaling pathway is critical for virtually all developmental programs and modulates an astounding variety of pathogenic processes. The DSL (Delta, Serrate, LAG-2 family) proteins have long been considered canonical activators of the core Notch pathway. More recently, a wide and expanding network of non-canonical extracellular factors has also been shown to modulate Notch signaling, conferring newly appreciated complexity to this evolutionarily conserved signal transduction system. Here, I review current concepts in Notch signaling, with a focus on work from the last decade elucidating novel extracellular proteins that up- or down-regulate signal potency.
Collapse
Affiliation(s)
- Michael M Wang
- Neurology Service, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA.
| |
Collapse
|
35
|
Duering M, Karpinska A, Rosner S, Hopfner F, Zechmeister M, Peters N, Kremmer E, Haffner C, Giese A, Dichgans M, Opherk C. Co-aggregate formation of CADASIL-mutant NOTCH3: a single-particle analysis. Hum Mol Genet 2011; 20:3256-65. [PMID: 21628316 DOI: 10.1093/hmg/ddr237] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
CADASIL (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy) is the most common monogenic cause of stroke and vascular dementia. Accumulation and deposition of the NOTCH3 (N3) extracellular domain in small blood vessels has been recognized as a central pathological feature of the disease. Recent experiments suggested enhanced formation of higher order multimers for mutant N3 compared with wild-type (WT). However, the mechanisms and consequences of N3 multimerization are still poorly understood, in part because of the lack of an appropriate in vitro aggregation assay. We therefore developed and validated a robust assay based on recombinant N3 fragments purified from cell culture supernatants. Using single-molecule analysis techniques such as scanning for intensely fluorescent targets and single-particle fluorescence resonance energy transfer, we show that spontaneous aggregation is limited to CADASIL-mutant N3, recapitulating a central aspect of CADASIL pathology in vitro. N3 aggregation requires no co-factor and is facilitated by sulfhydryl crosslinking. Although WT N3 does not exhibit multimerization itself, it can participate in aggregates of mutant N3. Furthermore, we demonstrate that thrombospondin-2, a known interaction partner of N3, co-aggregates with mutant N3. Sequestration of WT N3 and other proteins into aggregates represents a potentially important disease mechanism. These findings in combination with a new assay for single-molecule aggregation analysis provide novel opportunities for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Marco Duering
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University, 81377 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Meng H, Chen G, Zhang X, Wang Z, Thomas DG, Giordano TJ, Beer DG, Wang MM. Stromal LRP1 in lung adenocarcinoma predicts clinical outcome. Clin Cancer Res 2011; 17:2426-33. [PMID: 21325077 DOI: 10.1158/1078-0432.ccr-10-2385] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE LRP1 (low-density lipoprotein receptor-related protein 1) is a broadly expressed receptor that binds multiple extracellular ligands and participates in protein clearance. It is expressed in numerous cancers, but its role in lung cancer has not been characterized. Here, we investigate the relationship between LRP1 and lung cancer. EXPERIMENTAL DESIGN LRP1 mRNA levels were determined in lung tumors from several large, multicenter studies. LRP1 protein localization was determined by immunohistochemical analysis of lung tumor microarrays. Normal fibroblasts, fibroblasts treated with the LRP1 inhibitor RAP (receptor-associated protein), and Lrp1 null fibroblasts were cocultured with 3 independent lung cancer cell lines to investigate the role of LRP1 on tumor cell proliferation. RESULTS LRP1 mRNA levels are significantly decreased in lung tumors relative to nontumorous lung tissue. Lower expression of LRP1 in lung adenocarcinomas correlates with less favorable clinical outcome in a cohort of 439 patients. Immunohistochemical analysis shows that LRP1 is primarily expressed in stromal cells in 94/111 lung cancers, with very little protein found in cancer cells. A growth-suppressive function of mouse embryonic fibroblast (MEF) cells was observed in 3 lung cancer cell lines tested (H460, H2347, and HCC4006 cells); growth suppression was blocked by the LRP1 inhibitor RAP. Lrp1 deletion in fibroblasts reduced the ability of MEF cells to suppress tumor cell mitosis. In a validation set of adenocarcinomas, we confirmed a significant, positive correlation between both LRP1 mRNA and protein levels and favorable clinical outcomes. CONCLUSIONS LRP1 expression is associated with improved lung cancer outcomes. Mechanistically, stromal LRP1 may non-cell autonomously suppress lung tumor cell proliferation.
Collapse
Affiliation(s)
- He Meng
- Departments of Neurology, University of Michigan; and Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | |
Collapse
|