1
|
Li JS, Riggins K, Yang L, Chen C, Castro P, Alfarkh W, Zarrin-Khameh N, Scheurer ME, Creighton CJ, Musher B, Li W, Shen L. DNA methylation profiling at base-pair resolution reveals unique epigenetic features of early-onset colorectal cancer in underrepresented populations. Clin Epigenetics 2025; 17:11. [PMID: 39844333 PMCID: PMC11753045 DOI: 10.1186/s13148-025-01817-z] [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: 07/13/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND The incidence of early-onset colorectal cancer (EOCRC) has been rising at an alarming rate in the USA, and EOCRC disproportionately affects racial/ethnic minorities. Here, we construct comprehensive profiles of EOCRC DNA methylomes at base-pair resolution for a cohort of Hispanic and African American patients. RESULTS We show the epigenetic landscape of these EOCRC patients differs from that of late-onset colorectal cancer patients, and methylation canyons in EOCRC tumor tissue preferentially overlapped genes in cancer-related pathways. Furthermore, we identify epigenetic alterations in metabolic genes that are specific to our racial/ethnic minority EOCRC cohort but not Caucasian patients from TCGA. Top genes differentially methylated between these cohorts included the obesity-protective MFAP2 gene as well as cancer risk susceptibility genes APOL3 and RNASEL. CONCLUSIONS In this study, we provide to the scientific community high-resolution DNA methylomes for a cohort of EOCRC patients from underrepresented populations. Our exploratory findings in this cohort highlight epigenetic mechanisms underlying the pathogenesis of EOCRC and nominate novel biomarkers for EOCRC in underrepresented populations.
Collapse
Affiliation(s)
- Jason Sheng Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Karen Riggins
- Department of Medicine, Hematology and Oncology, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Li Yang
- Department of Pediatrics, USDA Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chaorong Chen
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Patricia Castro
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wedad Alfarkh
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Neda Zarrin-Khameh
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pathology, Ben Taub Hospital, 1504 Taub Loop, Houston, TX, 77030, USA
| | - Michael E Scheurer
- Department of Pediatrics, Center for Epidemiology and Population Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chad J Creighton
- Department of Medicine and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Benjamin Musher
- Department of Medicine, Gastrointestinal Medical Oncology, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA.
| | - Lanlan Shen
- Department of Pediatrics, USDA Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Dong SS, Duan YY, Zhu RJ, Jia YY, Chen JX, Huang XT, Tang SH, Yu K, Shi W, Chen XF, Jiang F, Hao RH, Liu Y, Liu Z, Guo Y, Yang TL. Systematic functional characterization of non-coding regulatory SNPs associated with central obesity. Am J Hum Genet 2025; 112:116-134. [PMID: 39753113 PMCID: PMC11739881 DOI: 10.1016/j.ajhg.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/03/2024] [Accepted: 11/13/2024] [Indexed: 01/20/2025] Open
Abstract
Central obesity is associated with higher risk of developing a wide range of diseases independent of overall obesity. Genome-wide association studies (GWASs) have identified more than 300 susceptibility loci associated with central obesity. However, the functional understanding of these loci is limited by the fact that most loci are in non-coding regions. To address this issue, our study first prioritized 2,034 single-nucleotide polymorphisms (SNPs) based on fine-mapping and epigenomic annotation analysis. Subsequently, we employed self-transcribing active regulatory region sequencing (STARR-seq) to systematically evaluate the enhancer activity of these prioritized SNPs. The resulting data analysis identified 141 SNPs with allelic enhancer activity. Further analysis of allelic transcription factor (TF) binding prioritized 20 key TFs mediating the central-obesity-relevant genetic regulatory network. Finally, as an example, we illustrate the molecular mechanisms of how rs8079062 acts as an allele-specific enhancer to regulate the expression of its targeted RNF157. We also evaluated the role of RNF157 in the adipogenic differentiation process. In conclusion, our results provide an important resource for understanding the genetic regulatory mechanisms underlying central obesity.
Collapse
Affiliation(s)
- Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yuan-Yuan Duan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Ren-Jie Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ying-Ying Jia
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Jia-Xin Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Xiao-Ting Huang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Shi-Hao Tang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ke Yu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Wei Shi
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Xiao-Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Feng Jiang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ruo-Han Hao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Zhongbo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| |
Collapse
|
3
|
Patel S, Ganbold K, Cho CH, Siddiqui J, Yildiz R, Sparman N, Sadeh S, Nguyen CM, Wang J, Whitelegge JP, Fried SK, Waki H, Villanueva CJ, Seldin MM, Sakaguchi S, Ellmeier W, Tontonoz P, Rajbhandari P. Transcription factor PATZ1 promotes adipogenesis by controlling promoter regulatory loci of adipogenic factors. Nat Commun 2024; 15:8533. [PMID: 39358382 PMCID: PMC11447024 DOI: 10.1038/s41467-024-52917-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
White adipose tissue (WAT) is essential for lipid storage and systemic energy homeostasis. Understanding adipocyte formation and stability is key to developing therapies for obesity and metabolic disorders. Through a high-throughput cDNA screen, we identified PATZ1, a POZ/BTB and AT-Hook Containing Zinc Finger 1 protein, as an important adipogenic transcription factor. PATZ1 is expressed in human and mouse adipocyte precursor cells (APCs) and adipocytes. In cellular models, PATZ1 promotes adipogenesis via protein-protein interactions and DNA binding. PATZ1 ablation in mouse adipocytes and APCs leads to a reduced APC pool, decreased fat mass, and hypertrophied adipocytes. ChIP-Seq and RNA-seq analyses show that PATZ1 supports adipogenesis by interacting with transcriptional machinery at the promoter regions of key early adipogenic factors. Mass-spec results show that PATZ1 associates with GTF2I, with GTF2I modulating PATZ1's function during differentiation. These findings underscore PATZ1's regulatory role in adipocyte differentiation and adiposity, offering insights into adipose tissue development.
Collapse
Affiliation(s)
- Sanil Patel
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Khatanzul Ganbold
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chung Hwan Cho
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Juwairriyyah Siddiqui
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ramazan Yildiz
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Njeri Sparman
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Shani Sadeh
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Christy M Nguyen
- Department of Biological Chemistry, University of California, Irvine, CA, 92697, USA
| | - Jiexin Wang
- Department of Pathology and Laboratory Medicine and Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
| | - Julian P Whitelegge
- Pasarow Mass Spectrometry Laboratory, NPI-Semel Institute, University of California, Los Angeles, CA, 90095, USA
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hironori Waki
- Department of Metabolism and Endocrinology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Claudio J Villanueva
- Molecular, Cellular, and Integrative Physiology Program, and Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, CA, 92697, USA
| | - Shinya Sakaguchi
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine and Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
4
|
Lv F, Li X, Wang Y, Hao L. MAGP1 maintains tumorigenicity and angiogenesis of laryngeal cancer by activating Wnt/β-catenin/MMP7 pathway. Carcinogenesis 2024; 45:220-234. [PMID: 36645203 DOI: 10.1093/carcin/bgad003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/17/2022] [Accepted: 01/16/2023] [Indexed: 01/17/2023] Open
Abstract
Microfibril-associated glycoprotein-1 (MAGP1), a crucial extracellular matrix protein, contributes to the initiation and progression of different cancers. However, the role of MAGP1 in laryngeal cancer is not clear. The purpose of this study was to investigate the clinical significance and biological function of MAGP1 in laryngeal cancer. MAGP1 was upregulated in public databases and laryngeal cancer tissues, and high MAGP1 expression led to a poor prognosis and was identified as an independent prognostic marker. Knocking-down MAGP1 inhibited laryngeal cancer cell growth and metastasis. According to gene set enrichment analysis, high MAGP1 expression revealed enrichment in Wnt/β-catenin signaling and knocking-down MAGP1 in laryngeal cancer cells also caused degradation, de-activation, re-location and loss of stability of β-catenin. Additionally, we observed MAGP1 in laryngeal cancer cells inhibits angiogenesis in an MMP7-dependent way. In conclusion, our study suggests a clinical role of MAGP1 in laryngeal cancer, signifying its potential as a therapeutic target in the future.
Collapse
Affiliation(s)
- Fei Lv
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaoqi Li
- Oncology Department III, People's Hospital of Liaoning Province, Shenyang, Liaoning, China
| | - Ying Wang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
6
|
Sun Y, Chen X, Chen L, Bao B, Li C, Zhou Y. MFAP2 promotes HSCs activation through FBN1/TGF-β/Smad3 pathway. J Cell Mol Med 2023; 27:3235-3246. [PMID: 37635348 PMCID: PMC10623529 DOI: 10.1111/jcmm.17884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/29/2023] Open
Abstract
Liver fibrosis is a chronic inflammatory process characterized by the accumulation of extracellular matrix (ECM), which contributes to cirrhosis and hepatocellular carcinoma. Increasing evidence suggests that the activation of hepatic stellate cells (HSCs) under an inflammatory state leads to the secretion of collagens, which can cause cirrhosis. In this study, we analysed data from the Gene Expression Omnibus (GEO) databases to identify differentially expressed genes (DEGs) between quiescent and fibrotic HSCs. We found that Microfibril Associated Protein 2 (MFAP2) was elevated in carbon tetrachloride (CCl4)-induced liver fibrosis and Transforming Growth Factor-Beta 1 (TGF-β1)-activated HSCs. Knockdown of MFAP2 inhibited HSC proliferation and partially attenuated TGF-β-stimulated fibrogenesis markers. Bioinformatics analysis revealed that Fibrillin-1 (FBN1) was correlated with MFAP2, and the expression of FBN1 was significantly upregulated after MFAP2 overexpression. Silencing MFAP2 partially attenuated the activation of HSCs by inhibiting HSC proliferation and decreasing collagen deposits. In vitro results showed that the inhibition of MFAP2 alleviated hepatic fibrosis by inhibiting the activation and inducing the apoptosis of active HSCs in a CCl4-induced mouse model. In conclusion, our results suggest that MFAP2 is a potential target for the clinical treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yonghong Sun
- Department of GastroenterologyThe First Hospital of Lanzhou UniversityLanzhouChina
- Department of PediatricsGansu Province People's HospitalLanzhouPeople's Republic of China
| | - Xingxing Chen
- Department of PediatricsGansu Province People's HospitalLanzhouPeople's Republic of China
| | - Lili Chen
- The First School of Clinical MedicineGansu University of Chinese MedicineLanzhouPeople's Republic of China
| | - Baixin Bao
- The First School of Clinical MedicineGansu University of Chinese MedicineLanzhouPeople's Republic of China
| | - Chunming Li
- Department of ObstetricsGansu Province People's HospitalLanzhouPeople's Republic of China
| | - Yongning Zhou
- Department of GastroenterologyThe First Hospital of Lanzhou UniversityLanzhouChina
| |
Collapse
|
7
|
Pan Y, Liu Z, Zhang H. Research progress of lens zonules. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2023; 3:80-85. [PMID: 37846380 PMCID: PMC10577871 DOI: 10.1016/j.aopr.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/23/2023] [Accepted: 02/23/2023] [Indexed: 10/18/2023]
Abstract
Background The lens zonule, a circumferential system of fibres connecting the ciliary body to the lens, is responsible for centration of the lens. The structural, functional, and positional abnormalities of the zonular apparatus can lead to the abnormality of the intraocular structure, presenting a significant challenge to cataract surgery. Main text The lens zonule is the elaborate system of extracellular fibers, which not only centers the lens in the eye but also plays an important role in accommodation and lens immunity, maintains the shape of the lens, and corrects spherical aberration. The zonules may directly participate in the formation of cataract via the immune mechanism. Abnormal zonular fibers that affect the position and shape of the lens may play an important role in the pathogenesis of angle closure disease and increase the complexity of the surgery. Capsular tension rings and related endocapsular devices are used to provide sufficient capsular bag stabilization and ensure the safety of cataract surgery procedures. Better preoperative and intraoperative evaluation methods for zonules are needed for clinicians. Conclusions The microstructure, biomechanical properties, and physiological functions of the lens zonules help us to better understand the pathogenesis of cataract and glaucoma, facilitating the development of safer surgical procedures for cataract. Further studies are needed to carefully analyze the structure-function relationship of the zonular apparatus to explore new treatment strategies for cataract and glaucoma.
Collapse
Affiliation(s)
- Yingying Pan
- Department of Ophthalmology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Zhaoqiang Liu
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Han Zhang
- Department of Ophthalmology, Shandong Provincial Hospital, Shandong University, Jinan, China
| |
Collapse
|
8
|
Microfibril-associated protein 2 is activated by POU class 2 homeobox 1 and promotes tumor growth and metastasis in tongue squamous cell carcinoma. Hum Cell 2023; 36:822-834. [PMID: 36527580 DOI: 10.1007/s13577-022-00840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022]
Abstract
Tongue squamous cell carcinoma (TSCC) represents the most frequent malignancy of the oral cavity, characterized by a high metastasis rate and poor prognosis. Microfibril-associated protein 2 (MFAP2), as an extracellular matrix protein, has been found to drive tumor progression. The function and underlying mechanism of MFAP2 in TSCC remain unknown. The expression levels of MFAP2 were analyzed in tissue samples from 30 TSCC patients by real time-polymerase chain reaction and western blot assays. Our results revealed that the expression of MFAP2 mRNA and protein was upregulated in TSCC tissue samples compared with that in the matched para-carcinoma tissue samples. By performing in vitro gain-of-function or loss-of-function experiments and in vivo mouse xenograft experiments, we found that overexpression of MFAP2 induced proliferation and promoted transition from G1 to S phase of TSCC cells. Stronger invasive and migratory capabilities were observed in MFAP2-overexpressing TSCC cells. In contrast, knockdown of MFAP2 exhibited anti-proliferative, apoptosis-promoting and pro-migratory roles in TSCC cells. Knockdown of MFAP2 significantly inhibited xenograft tumor growth. Mechanistically, POU class 2 homeobox 1 (POU2F1) was recruited to the region of MFAP2 promoter and upregulates the expression of MFAP2. Silencing of MFAP2 effectively blocked the proliferation, migration, and invasion of TSCC cells caused by POU2F1 overexpression. Our results indicate that the role of MFAP2 in TSCC may attribute to transcriptional regulation of POU2F1.
Collapse
|
9
|
Pietka TA, Abumrad NA. Determination of Adipocyte Size and Number. Methods Mol Biol 2023; 2662:33-52. [PMID: 37076669 DOI: 10.1007/978-1-0716-3167-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Dysfunction in adipocyte expansion during the onset of obesity is associated with metabolic abnormalities. Determination of adipocyte size and number is an important measure for a comprehensive evaluation of the metabolic status of adipose tissue. Here, we describe three methods for the determination of adipocyte size that can be applied to tissue samples obtained from humans and rodent models. While the first method presented is more robust, it does require the use of osmium, a toxic heavy metal, which requires special handling and disposal precautions in addition to specialized equipment. Two additional methods are described that can be of use to most researchers.
Collapse
Affiliation(s)
- Terri A Pietka
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
- Cellular and Molecular Biology Core, Washington University Nutrition and Obesity Research Center, St Louis, MO, USA.
| | - Nada A Abumrad
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Cellular and Molecular Biology Core, Washington University Nutrition and Obesity Research Center, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
10
|
Xu W, Wang M, Bai Y, Chen Y, Ma X, Yang Z, Zhao L, Li Y. The role of microfibrillar‐associated protein 2 in cancer. Front Oncol 2022; 12:1002036. [DOI: 10.3389/fonc.2022.1002036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/03/2022] [Indexed: 12/05/2022] Open
Abstract
Microfibrillar-associated protein 2 (MFAP2), a component of the extracellular matrix, is important in controlling growth factor signal transduction. Recent studies have shown that MFAP2, an effective prognostic molecule for various tumors, is associated with tumor occurrence and development and may be involved in remodeling the extracellular matrix and regulating proliferation, apoptosis, invasion, tumor cell metastasis, and tumor angiogenesis. However, MFAP2’s specific mechanism in these tumor processes remains unclear. This article reviewed the possible mechanism of MFAP2 in tumorigenesis and progression and provided a reference for the clinical prognosis of patients with cancer and new therapeutic target discovery.
Collapse
|
11
|
Singer JM, Shew TM, Ferguson D, Renkemeyer MK, Pietka TA, Hall AM, Finck BN, Lutkewitte AJ. Monoacylglycerol O-acyltransferase 1 lowers adipocyte differentiation capacity in vitro but does not affect adiposity in mice. Obesity (Silver Spring) 2022; 30:2122-2133. [PMID: 36321276 PMCID: PMC9634674 DOI: 10.1002/oby.23538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Monoacylglycerol O-acyltransferase 1 (Mogat1), a lipogenic enzyme that converts monoacylglycerol to diacylglycerol, is highly expressed in adipocytes and may regulate lipolysis by re-esterifying fatty acids released during times when lipolytic rates are low. However, the role of Mogat1 in regulating adipocyte fat storage during differentiation and diet-induced obesity is relatively understudied. METHODS Here, adipocyte-specific Mogat1 knockout mice were generated and subjected to a high-fat diet to determine the effects of Mogat1 deficiency on diet-induced obesity. Mogat1 floxed mice were also used to develop preadipocyte cell lines wherein Mogat1 could be conditionally knocked out to study adipocyte differentiation in vitro. RESULTS In preadipocytes, it was found that Mogat1 knockout at the onset of preadipocyte differentiation prevented the accumulation of glycerolipids and reduced the differentiation capacity of preadipocytes. However, the loss of adipocyte Mogat1 did not affect weight gain or fat mass induced by a high-fat diet in mice. Furthermore, loss of Mogat1 in adipocytes did not affect plasma lipid or glucose concentrations or insulin tolerance. CONCLUSIONS These data suggest Mogat1 may play a role in adipocyte differentiation in vitro but not adipose tissue expansion in response to nutrient overload in mice.
Collapse
Affiliation(s)
- Jason M. Singer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Trevor M. Shew
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Daniel Ferguson
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - M. Katie Renkemeyer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Terri A. Pietka
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Angela M. Hall
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Brian N. Finck
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Andrew J. Lutkewitte
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
12
|
Singh K, Sachan N, Ene T, Dabovic B, Rifkin D. Latent Transforming Growth Factor β Binding Protein 3 Controls Adipogenesis. Matrix Biol 2022; 112:155-170. [PMID: 35933071 DOI: 10.1016/j.matbio.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022]
Abstract
Transforming growth factor-beta (TGFβ) is released from cells as part of a trimeric latent complex consisting of TGFβ, the TGFβ propeptides, and either a latent TGFβ binding protein (LTBP) or glycoprotein-A repetitions predominant (GARP) protein. LTBP1 and 3 modulate latent TGFβ function with respect to secretion, matrix localization, and activation and, therefore, are vital for the proper function of the cytokine in a number of tissues. TGFβ modulates stem cell differentiation into adipocytes (adipogenesis), but the potential role of LTBPs in this process has not been studied. We observed that 72 h post adipogenesis initiation Ltbp1, 2, and 4 expression levels decrease by 74-84%, whereas Ltbp3 expression levels remain constant during adipogenesis. We found that LTBP3 silencing in C3H/10T1/2 cells reduced adipogenesis, as measured by the percentage of cells with lipid vesicles and the expression of the transcription factor peroxisome proliferator-activated receptor gamma (PPARγ). Lentiviral mediated expression of an Ltbp3 mRNA resistant to siRNA targeting rescued the phenotype, validating siRNA specificity. Knockdown (KD) of Ltbp3 expression in 3T3-L1, M2, and primary bone marrow stromal cells (BMSC) indicated a similar requirement for Ltbp3. Epididymal and inguinal white adipose tissue fat pad weights of Ltbp3-/- mice were reduced by 62% and 57%, respectively, compared to wild-type mice. Inhibition of adipogenic differentiation upon LTBP3 loss is mediated by TGFβ, as TGFβ neutralizing antibody and TGFβ receptor I kinase blockade rescue the LTBP3 KD phenotype. These results indicate that LTBP3 has a TGFβ-dependent function in adipogenesis both in vitro and possibly in vivo.
Collapse
Affiliation(s)
- Karan Singh
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nalani Sachan
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Taylor Ene
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Branka Dabovic
- Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Daniel Rifkin
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA; Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Muthu ML, Tiedemann K, Fradette J, Komarova S, Reinhardt DP. Fibrillin-1 regulates white adipose tissue development, homeostasis, and function. Matrix Biol 2022; 110:106-128. [DOI: 10.1016/j.matbio.2022.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/12/2022] [Accepted: 05/04/2022] [Indexed: 12/28/2022]
|
14
|
Pan-Cancer Analysis of Microfibrillar-Associated Protein 2 (MFAP2) Based on Bioinformatics and qPCR Verification. JOURNAL OF ONCOLOGY 2022; 2022:8423173. [PMID: 35211173 PMCID: PMC8863482 DOI: 10.1155/2022/8423173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 11/18/2022]
Abstract
MFAP2 has been reported to play an oncogenic role in several types of human cancers. However, the expression profile of MFAP2 in various cancers and its impact on prognosis and immune infiltration remain unclear. In this study, the mRNA expression and protein expression of MFAP2 in normal tissues, tumor cell lines, and 33 malignant tumor tissues were analyzed comprehensively using Genotype-Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), and The Cancer Genome Atlas (TCGA), Oncomine and UALCAN databases, and the expression of MFAP2 in different grades and stages of cancers was assessed using Gene Expression Profiling Interactive Analysis 2 (GEPIA2) and Tumor and Immune System Interaction Database (TISIDB). In general, MFAP2 showed distinct expression in most tumor and normal tissues, closely associated with higher tumor grade, higher tumor stage, and poor survival in multiple cancers. A search of the UALCAN database and the cBioPortal database revealed that this difference in mRNA level expression could be partly attributed to abnormal DNA methylation and mutations at the genomic level. In addition, MFAP2 expression was also associated with tumor mutation burden, microsatellite instability, and neoantigens in different cancer types. More importantly, the TIMER and TISIDB databases also showed that MFAP2 levels were significantly correlated with immune infiltration abundance and immune-related gene markers, as well as ESTIMATE scores. By qPCR, MFAP2 expression was validated in four kinds of tumor tissue samples. The present study combined several databases and performed a pan-cancer analysis of the expression profile, methylation, and mutation for MFAP2 and its implications for prognosis and immune infiltration, suggesting that MFAP2 could contribute to malignant properties of many tumors. MFAP2 may be an important biomarker with prognostic value and has the potential to be a target for tumor immunotherapy.
Collapse
|
15
|
Kong J, Zhou X, Lu J, Han Q, Ouyang X, Chen D, Liu A. Maclurin Promotes the Chondrogenic Differentiation of Bone Marrow Mesenchymal Stem Cells by Regulating miR-203a-3p/Smad1. Cell Reprogram 2022; 24:9-20. [PMID: 35180001 DOI: 10.1089/cell.2021.0122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) differentiate into chondrocytes under appropriate conditions, providing a method for the treatment of bone- and joint-related diseases. Previously, we found that mulberry (Morus nigra) promoted the chondrogenic differentiation of BMSCs. Although the mechanism of action and active ingredients remain unknown, several studies describe the involvement of micro-RNAs. We obtained BMSCs from the bone marrow of Sprague Dawley rats. Cell Counting Kit-8 assays showed that maclurin (25 μg/mL) treatment was not toxic to BMSCs, and compared with untreated controls, maclurin upregulated Sox9 and Col2a expression. Quantitative-PCR revealed that miR-203a-3p levels decreased significantly during chondrogenic differentiation of BMSCs promoted by maclurin. Compared with treatment with an miR-203a-3p inhibitor, miR-203a-3p mimic inhibited expression of Sox9 and Col2a as evidenced by immunofluorescence staining and Western blotting. Smad1 was identified as a key target gene of miR-203a-3p according to biological-prediction software, and miR-203a-3p negatively regulated its transcription and translation in the dual-luciferase reporter gene assay and Western blotting. Sox9 and Col2a expression was downregulated following transfection of short interfering Smad1 (siSmad1) plasmids into BMSCs. We elucidated how maclurin promotes the chondrogenic differentiation of BMSCs by regulating miR-203a-3p/Smad1, which provides a strategy for future exploration of osteoarthritis therapy through cell transplantation.
Collapse
Affiliation(s)
- Jiechen Kong
- Center for Experimental Teaching, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xianxi Zhou
- Center for Experimental Teaching, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianghua Lu
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qianting Han
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiyan Ouyang
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dongfeng Chen
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Aijun Liu
- Center for Experimental Teaching, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Gómez de Segura I, Ahechu P, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Becerril S, Unamuno X, Mentxaka A, Baixauli J, Valentí V, Moncada R, Silva C, Frühbeck G, Catalán V. Decreased Levels of Microfibril-Associated Glycoprotein (MAGP)-1 in Patients with Colon Cancer and Obesity Are Associated with Changes in Extracellular Matrix Remodelling. Int J Mol Sci 2021; 22:ijms22168485. [PMID: 34445187 PMCID: PMC8395192 DOI: 10.3390/ijms22168485] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE The protein microfibril-associated glycoprotein (MAGP)-1 constitutes a crucial extracellular matrix protein. We aimed to determine its impact on visceral adipose tissue (VAT) remodelling during obesity-associated colon cancer (CC). METHODS Samples obtained from 79 subjects (29 normoponderal (NP) (17 with CC) and 50 patients with obesity (OB) (19 with CC)) were used in the study. Circulating concentrations of MAGP-1 and its gene expression levels (MFAP2) in VAT were analysed. The impact of inflammation-related factors and adipocyte-conditioned media (ACM) on MFAP2 mRNA levels in colon adenocarcinoma HT-29 cells were further analysed. The effects of MAGP-1 in the expression of genes involved in the extracellular matrix (ECM) remodelling and tumorigenesis in HT-29 cells was also explored. RESULTS Obesity (p < 0.01) and CC (p < 0.001) significantly decreased MFAP2 gene expression levels in VAT whereas an opposite trend in TGFB1 mRNA levels was observed. Increased mRNA levels of MFAP2 after the stimulation of HT-29 cells with lipopolysaccharide (LPS) (p < 0.01) and interleukin (IL)-4 (p < 0.01) together with a downregulation (p < 0.05) after hypoxia mimicked by CoCl2 treatment was observed. MAGP-1 treatment significantly enhanced the mRNA levels of the ECM-remodelling genes collagen type 6 α3 chain (COL6A3) (p < 0.05), decorin (DCN) (p < 0.01), osteopontin (SPP1) (p < 0.05) and TGFB1 (p < 0.05). Furthermore, MAGP-1 significantly reduced (p < 0.05) the gene expression levels of prostaglandin-endoperoxide synthase 2 (COX2/PTGS2), a key gene controlling cell proliferation, growth and adhesion in CC. Interestingly, a significant decrease (p < 0.01) in the mRNA levels of MFAP2 in HT-29 cells preincubated with ACM from volunteers with obesity compared with control media was observed. Conclusion: The decreased levels of MAGP-1 in patients with obesity and CC together with its capacity to modulate key genes involved in ECM remodelling and tumorigenesis suggest MAGP-1 as a link between AT excess and obesity-associated CC development.
Collapse
Affiliation(s)
- Iranzu Gómez de Segura
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
| | - Patricia Ahechu
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (P.A.); (J.B.); (V.V.)
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
| | - Amaia Mentxaka
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
| | - Jorge Baixauli
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (P.A.); (J.B.); (V.V.)
| | - Víctor Valentí
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (P.A.); (J.B.); (V.V.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Rafael Moncada
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Department of Anesthesia, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Camilo Silva
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Correspondence: (G.F.); (V.C.); Tel.: +34-9-4825-5400 (ext. 4484) (G.F.); +34-9-4825-5400 (ext. 5133) (V.C.)
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
- Correspondence: (G.F.); (V.C.); Tel.: +34-9-4825-5400 (ext. 4484) (G.F.); +34-9-4825-5400 (ext. 5133) (V.C.)
| |
Collapse
|
17
|
The effect of TGFβ1 on thermogenic markers is dependent on the degree of adipocyte differentiation. Biosci Rep 2021; 40:223097. [PMID: 32352511 PMCID: PMC7225410 DOI: 10.1042/bsr20194262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 12/02/2022] Open
Abstract
Transforming growth factor β (TGFβ) a multifunctional cytokine is known to regulate cell proliferation, differentiation, migration and survival. Although there is variable expression of modulators of TGFβ action during differentiation, a differential effect on fat cell metabolism at the different stages of adipocyte differentiation was unclear. In the present study, 3T3L1 cells were used as an in vitro model to study the effect of TGFβ on adipogenic and thermogenic markers at various stages of preadipocyte to mature adipocyte differentiation. As in our earlier studies on the effect of TGFβ on CEBP’s, we used a standard differentiation mix, and one with the addition of rosiglitazone. RhTGFβ1 was added to undifferentiated adipocytes (preadipocytes) and to adipocytes at day 0 (commitment stage) as well as day 10 (terminal differentiation). Cellular responses in terms of Pref1, PPARγ, TLE3, PGC1α, PRDM16, UCP1 and UCP2 mRNA levels and selected protein products, were determined. Increases in PPARγ, PRDM16, UCP1 and UCP2 mRNA and decreases in Pref1 are good indicators of successful differentiation. The early addition of rhTGFβ1 during commitment stage decreased PPARγ, PRDM16, TLE3, UCP1 and UCP2 mRNA and decreased PRDM16 protein consistent with our earlier report on the inhibition of CEBP’s by TGFβ and CCN2. The addition of rhTGFβ1 to mature adipocyte at day 10 increased UCP1 mRNA and increased PRDM16 and UCP1 proteins. In the present study, our results suggest that TGFβ1 added late enhances the thermogenic potential of mature cells and causes 3T3L1 cells to differentiate to resemble brown or beige rather than white adipose tissue.
Collapse
|
18
|
Zhu S, Ye L, Bennett S, Xu H, He D, Xu J. Molecular structure and function of microfibrillar-associated proteins in skeletal and metabolic disorders and cancers. J Cell Physiol 2020; 236:41-48. [PMID: 32572962 DOI: 10.1002/jcp.29893] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/16/2022]
Abstract
Microfibrillar-associated proteins (MFAPs) are extracellular matrix glycoproteins, which play a role in microfibril assembly, elastinogenesis, and tissue homeostasis. MFAPs consist of five subfamily members, including MFAP1, MFAP2, MFAP3, MFAP4, and MFAP5. Among these, MFAP2 and MFAP5 are most closely related, and exhibit very limited amino acid sequence homology with MFAP1, MFAP3, and MFAP4. Gene expression profiling analysis reveals that MFAP2, MFAP5, and MFAP4 are specifically expressed in osteoblastic like cells, whereas MFAP1 and MFAP3 are more ubiquitously expressed, indicative of their diverse role in the tropism of tissues. Molecular structural analysis shows that each MFAP family member has distinct features, and functional evidence reveals discrete purposes of individual MFAPs. Animal studies indicate that MFAP2-deficient mice exhibit progressive osteopenia with elevated receptor activator of NF-κB ligand (RANKL) expression, whereas MFAP5-deficient mice are neutropenic, and MFAP4-deficient mice displayed emphysema-like pathology and the impaired formation of neointimal hyperplasia. Emerging data also suggest that MFAPs are involved in cancer progression and fat metabolism. Further understanding of tissue-specific pathophysiology of MFAPs might offer potential novel therapeutic targets for related diseases, such as skeletal and metabolic disorders, and cancers.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Lin Ye
- Department of Orthopaedic Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Samuel Bennett
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dengwei He
- Department of Orthopaedic Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| |
Collapse
|
19
|
Systems Genetics in Human Endothelial Cells Identifies Non-coding Variants Modifying Enhancers, Expression, and Complex Disease Traits. Am J Hum Genet 2020; 106:748-763. [PMID: 32442411 DOI: 10.1016/j.ajhg.2020.04.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/05/2020] [Indexed: 12/22/2022] Open
Abstract
The identification of causal variants and mechanisms underlying complex disease traits in humans is important for the progress of human disease genetics; this requires finding strategies to detect functional regulatory variants in disease-relevant cell types. To achieve this, we collected genetic and transcriptomic data from the aortic endothelial cells of up to 157 donors and four epigenomic phenotypes in up to 44 human donors representing individuals of both sexes and three major ancestries. We found thousands of expression quantitative trait loci (eQTLs) at all ranges of effect sizes not detected by the Gene-Tissue Expression Project (GTEx) in human tissues, showing that novel biological relationships unique to endothelial cells (ECs) are enriched in this dataset. Epigenetic profiling enabled discovery of over 3,000 regulatory elements whose activity is modulated by genetic variants that most frequently mutated ETS, AP-1, and NF-kB binding motifs, implicating these motifs as governors of EC regulation. Using CRISPR interference (CRISPRi), allele-specific reporter assays, and chromatin conformation capture, we validated candidate enhancer variants located up to 750 kb from their target genes, VEGFC, FGD6, and KIF26B. Regulatory SNPs identified were enriched in coronary artery disease (CAD) loci, and this result has specific implications for PECAM-1, FES, and AXL. We also found significant roles for EC regulatory variants in modifying the traits pulse pressure, blood protein levels, and monocyte count. Lastly, we present two unlinked SNPs in the promoter of MFAP2 that exhibit pleiotropic effects on human disease traits. Together, this supports the possibility that genetic predisposition for complex disease is manifested through the endothelium.
Collapse
|
20
|
DeDreu J, Bowen CJ, Logan CM, Pal-Ghosh S, Parlanti P, Stepp MA, Menko AS. An immune response to the avascular lens following wounding of the cornea involves ciliary zonule fibrils. FASEB J 2020; 34:9316-9336. [PMID: 32452112 PMCID: PMC7384020 DOI: 10.1096/fj.202000289r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022]
Abstract
The lens and central cornea are avascular. It was assumed that the adult lens had no source of immune cells and that the basement membrane capsule surrounding the lens was a barrier to immune cell migration. Yet, microfibril‐associated protein‐1 (MAGP1)‐rich ciliary zonules that originate from the vasculature‐rich ciliary body and extend along the surface of the lens capsule, form a potential conduit for immune cells to the lens. In response to cornea debridement wounding, we find increased expression of MAGP1 throughout the central corneal stroma. The immune cells that populate this typically avascular region after wounding closely associate with this MAGP1‐rich matrix. These results suggest that MAGP1‐rich microfibrils support immune cell migration post‐injury. Using this cornea wound model, we investigated whether there is an immune response to the lens following cornea injury involving the lens‐associated MAGP1‐rich ciliary zonules. Our results provide the first evidence that following corneal wounding immune cells are activated to travel along zonule fibers that extend anteriorly along the equatorial surface of the lens, from where they migrate across the anterior lens capsule. These results demonstrate that lens‐associated ciliary zonules are directly involved in the lens immune response and suggest the ciliary body as a source of immune cells to the avascular lens.
Collapse
Affiliation(s)
- JodiRae DeDreu
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Caitlin J Bowen
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Caitlin M Logan
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Paola Parlanti
- George Washington University Nanofabrication and Imaging Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.,Department of Ophthalmology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - A Sue Menko
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
21
|
Fibrillin-1 and fibrillin-1-derived asprosin in adipose tissue function and metabolic disorders. J Cell Commun Signal 2020; 14:159-173. [PMID: 32279186 DOI: 10.1007/s12079-020-00566-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/28/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix microenvironment of adipose tissue is of critical importance for the differentiation, remodeling and function of adipocytes. Fibrillin-1 is one of the main components of microfibrils and a key player in this process. Furin processing of profibrillin-1 results in mature fibrillin-1 and releases the C-terminal propeptide as a circulating hunger hormone, asprosin. Mutations in the fibrillin-1 gene lead to adipose tissue dysfunction and causes Marfan syndrome, marfanoid progeroid lipodystrophy syndrome, and neonatal progeroid syndrome. Increased TGF-β signaling, altered mechanical properties and impaired adipogenesis are potential causes of adipose tissue dysfunction, mediated through deficient microfibrils. Circulating asprosin on the other hand is secreted primarily by white adipose tissue under fasting conditions and in obesity. It increases hepatic glucose production and drives insulin secretion and appetite stimulation through inter-organ cross talk. This review discusses the metabolic consequences of fibrillin-1 and fibrillin-1-derived asprosin in pathological conditions. Understanding the dynamic role of fibrillin-1 in the adipose tissue milieu and of circulating asprosin in the body can provide novel mechanistic insights into how fibrillin-1 may contribute to metabolic syndrome. This could lead to new management regimens of patients with metabolic disease.
Collapse
|
22
|
Yao LW, Wu LL, Zhang LH, Zhou W, Wu L, He K, Ren JC, Deng YC, Yang DM, Wang J, Mu GG, Xu M, Zhou J, Xiang GA, Ding QS, Yang YN, Yu HG. MFAP2 is overexpressed in gastric cancer and promotes motility via the MFAP2/integrin α5β1/FAK/ERK pathway. Oncogenesis 2020; 9:17. [PMID: 32054827 PMCID: PMC7018958 DOI: 10.1038/s41389-020-0198-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 12/23/2019] [Accepted: 01/23/2020] [Indexed: 12/17/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies and its prognosis is extremely poor. This study identifies a novel oncogene, microfibrillar-associated protein 2 (MFAP2) in GC. With integrative reanalysis of transcriptomic data, we found MFAP2 as a GC prognosis-related gene. And the aberrant expression of MFAP2 was explored in GC samples. Subsequent experiments indicated that silencing and exogenous MFAP2 could affect motility of cancer cells. The inhibition of silencing MFAP2 could be rescued by another FAK activator, fibronectin. This process is probably through affecting the activation of focal adhesion process via modulating ITGB1 and ITGA5. MFAP2 regulated integrin expression through ERK1/2 activation. Silencing MFAP2 by shRNA inhibited tumorigenicity and metastasis in nude mice. We also revealed that MFAP2 is a novel target of microRNA-29, and miR-29/MFAP2/integrin α5β1/FAK/ERK1/2 could be an important oncogenic pathway in GC progression. In conclusion, our data identified MFAP2 as a novel oncogene in GC and revealed that miR-29/MFAP2/integrin α5β1/FAK/ERK1/2 could be an important oncogenic pathway in GC progression.
Collapse
Affiliation(s)
- Li-Wen Yao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Lian-Lian Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Li-Hui Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Wei Zhou
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Lu Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Ke He
- Department of General Surgery, The Second People's Hospital of Guangdong Province, Southern Medical University, Guangzhou, Guangdong, 510317, P.R. China
- Department of Biochemistry, Zhongshan Medical College, Sun Yat-sen University, Guangzhou, P.R. China
| | - Jia-Cai Ren
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Yun-Chao Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Dong-Mei Yang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Jing Wang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Gang-Gang Mu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Ming Xu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Jie Zhou
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Guo-An Xiang
- Department of General Surgery, The Second People's Hospital of Guangdong Province, Southern Medical University, Guangzhou, Guangdong, 510317, P.R. China
| | - Qian-Shan Ding
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China.
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China.
| | - Yan-Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China.
| | - Hong-Gang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China.
- Hubei Key laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China.
| |
Collapse
|
23
|
Marcelin G, Da Cunha C, Gamblin C, Suffee N, Rouault C, Leclerc A, Lacombe A, Sokolovska N, Gautier EL, Clément K, Dugail I. Autophagy inhibition blunts PDGFRA adipose progenitors' cell-autonomous fibrogenic response to high-fat diet. Autophagy 2020; 16:2156-2166. [PMID: 31992125 DOI: 10.1080/15548627.2020.1717129] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adipose tissue (AT) fibrosis in obesity compromises adipocyte functions and responses to intervention-induced weight loss. It is driven by AT progenitors with dual fibro/adipogenic potential, but pro-fibrogenic pathways activated in obesity remain to be deciphered. To investigate the role of macroautophagy/autophagy in AT fibrogenesis, we used Pdgfra-CreErt2 transgenic mice to create conditional deletion of Atg7 alleles in AT progenitor cells (atg7 cKO) and examined sex-dependent, depot-specific AT remodeling in high-fat diet (HFD)-fed mice. Mice with atg7 cKO had markedly decreased extracellular matrix (ECM) gene expression in visceral, subcutaneous, and epicardial adipose depots compared to Atg7lox/lox littermates. ECM gene program regulation by autophagy inhibition occurred independently of changes in the mass of fat tissues or adipocyte numbers of specific depots, and cultured preadipocytes treated with pharmacological or siRNA-mediated autophagy disruptors could mimic these effects. We found that autophagy inhibition promotes global cell-autonomous remodeling of the paracrine TGF-BMP family landscape, whereas ECM gene modulation was independent of the autophagic regulation of GTF2IRD1. The progenitor-specific mouse model of ATG7 inhibition confirms the requirement of autophagy for white/beige adipocyte turnover, and combined to in vitro experiments, reveal progenitor autophagy dependence for AT fibrogenic response to HFD, through the paracrine remodeling of TGF-BMP factors balance. Abbreviations: CQ: chloroquine; ECM: extracellular matrix; EpiAT: epididymal adipose tissue; GTF2IRD1: general transcription factor II I repeat domain-containing 1; HFD: high-fat diet; KO: knockout; OvAT: ovarian adipose tissue; PDGFR: platelet derived growth factor receptor; ScAT: subcutaneous adipose tissue; TGF-BMP: transforming growth factor-bone morphogenic protein.
Collapse
Affiliation(s)
| | - Carla Da Cunha
- UMRS1269 INSERM/Sorbonne University, Nutriomics , Paris, France
| | - Camille Gamblin
- UMRS1269 INSERM/Sorbonne University, Nutriomics , Paris, France
| | | | | | - Arnaud Leclerc
- UMRS1269 INSERM/Sorbonne University, Nutriomics , Paris, France
| | - Amelie Lacombe
- Institute of Cardiometabolism and Nutrition (ICAN) , Paris, France
| | | | | | - Karine Clément
- UMRS1269 INSERM/Sorbonne University, Nutriomics , Paris, France.,Pitié-Salpêtrière hospital, Nutrition department, Asssistance Publique-Hôpitaux de Paris , Paris, France
| | - Isabelle Dugail
- UMRS1269 INSERM/Sorbonne University, Nutriomics , Paris, France
| |
Collapse
|
24
|
Broekelmann TJ, Bodmer NK, Mecham RP. Identification of the growth factor-binding sequence in the extracellular matrix protein MAGP-1. J Biol Chem 2020; 295:2687-2697. [PMID: 31988245 DOI: 10.1074/jbc.ra119.010540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Microfibril-associated glycoprotein-1 (MAGP-1) is a component of vertebrate extracellular matrix (ECM) microfibrils that, together with the fibrillins, contributes to microfibril function. Many of the phenotypes associated with MAGP-1 gene inactivation are consistent with dysregulation of the transforming growth factor β (TGFβ)/bone morphogenetic protein (BMP) signaling system. We have previously shown that full-length MAGP-1 binds active TGFβ-1 and some BMPs. The work presented here further defines the growth factor-binding domain of MAGP-1. Using recombinant domains and synthetic peptides, along with surface plasmon resonance analysis to measure the kinetics of the MAGP-1-TGFβ-1 interaction, we localized the TGFβ- and BMP-binding site in MAGP-1 to a 19-amino acid-long, highly acidic sequence near the N terminus. This domain was specific for binding active, but not latent, TGFβ-1. Growth factor activity experiments revealed that TGFβ-1 retains signaling activity when complexed with MAGP-1. Furthermore, when bound to fibrillin, MAGP-1 retained the ability to interact with TGFβ-1, and active TGFβ-1 did not bind fibrillin in the absence of MAGP-1. The absence of MAGP was sufficient to raise the amount of total TGFβ stored in the ECM of cultured cells, suggesting that the MAGPs compete with the TGFβ large latent complex for binding to microfibrils. Together, these results indicate that MAGP-1 plays an active role in TGFβ signaling in the ECM.
Collapse
Affiliation(s)
- Thomas J Broekelmann
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Nicholas K Bodmer
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110.
| |
Collapse
|
25
|
Wu M, Ding Y, Jiang X, Chen Y, Wu N, Li L, Wang H, Huang Y, Xu N, Teng L. Overexpressed MAGP1 Is Associated With a Poor Prognosis and Promotes Cell Migration and Invasion in Gastric Cancer. Front Oncol 2020; 9:1544. [PMID: 32010630 PMCID: PMC6978879 DOI: 10.3389/fonc.2019.01544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/20/2019] [Indexed: 01/14/2023] Open
Abstract
Gastric cancer (GC) is a frequently occurring malignancy with high mortality rates. However, the underlying mechanism of GC progression is not very clear. The aim of this study is to reveal the inherent molecular mechanism and develop potential therapeutic targets for advanced GC. The microfibril-associated glycoprotein 1 (MAGP1), identified as a potential oncogene, was found upregulated in GC tissues and high MAGP1 expression was associated with aggressive clinicopathological features. Furthermore, the multivariate Cox regression analysis showed that high MAGP1 expression was an independent predictor of poor prognosis (HR = 2.37, 1.07-5.24; P = 0.033). Mechanistically, MAGP1 promoted the migration and invasiveness of GC cells. In addition, the genes co-expressed with MAGP1 were primarily enriched in focal adhesion and PI3K-Akt pathways. MAGP1 overexpression enhanced the phosphorylation of FAK, AKT, and mTOR, whereas its knockdown also inactivated these factors. Furthermore, the AKT inhibitor suppressed the phosphorylation of AKT, FAK, and mTOR in recMAGP1-treated AGS cells, as well as their migration and invasion capacities. Finally, correlation analysis indicated that MAGP1 is involved in AKT signaling in GC, and is clinically relevant. Taken together, MAGP1 is a promising prognostic marker and potential therapeutic target for advanced GC.
Collapse
Affiliation(s)
- Mengjie Wu
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yongfeng Ding
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxia Jiang
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanyan Chen
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nan Wu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linrong Li
- Department of Otorhinolaryngology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haiyong Wang
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yingying Huang
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nong Xu
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lisong Teng
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Wahart A, Hocine T, Albrecht C, Henry A, Sarazin T, Martiny L, El Btaouri H, Maurice P, Bennasroune A, Romier-Crouzet B, Blaise S, Duca L. Role of elastin peptides and elastin receptor complex in metabolic and cardiovascular diseases. FEBS J 2019; 286:2980-2993. [PMID: 30946528 DOI: 10.1111/febs.14836] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/23/2019] [Accepted: 04/02/2019] [Indexed: 12/11/2022]
Abstract
The Cardiovascular Continuum describes a sequence of events from cardiovascular risk factors to end-stage heart disease. It includes conventional pathologies affecting cardiovascular functions such as hypertension, atherosclerosis or thrombosis and was traditionally considered from the metabolic point of view. This Cardiovascular Continuum, originally described by Dzau and Braunwald, was extended by O'Rourke to consider also the crucial role played by degradation of elastic fibers, occurring during aging, in the appearance of vascular stiffness, another deleterious risk factor of the continuum. However, the involvement of the elastin degradation products, named elastin-derived peptides, to the Cardiovascular Continuum progression has not been considered before. Data from our laboratory and others clearly showed that these bioactive peptides are central regulators of this continuum, thereby amplifying appearance and evolution of cardiovascular risk factors such as diabetes or hypertension, of vascular alterations such as atherothrombosis and calcification, but also nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. The Elastin Receptor Complex has been shown to be a crucial actor in these processes. We propose here the participation of these elastin-derived peptides and of the Elastin Receptor Complex in these events, and introduce a revisited Cardiovascular Continuum based on their involvement, for which elastin-based pharmacological strategies could have a strong impact in the future.
Collapse
Affiliation(s)
- Amandine Wahart
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Thinhinane Hocine
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Camille Albrecht
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Auberi Henry
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Thomas Sarazin
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Laurent Martiny
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Hassan El Btaouri
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Amar Bennasroune
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | | | - Sébastien Blaise
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| |
Collapse
|
27
|
Stenvers DJ, Jongejan A, Atiqi S, Vreijling JP, Limonard EJ, Endert E, Baas F, Moerland PD, Fliers E, Kalsbeek A, Bisschop PH. Diurnal rhythms in the white adipose tissue transcriptome are disturbed in obese individuals with type 2 diabetes compared with lean control individuals. Diabetologia 2019; 62:704-716. [PMID: 30737520 DOI: 10.1007/s00125-019-4813-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/18/2018] [Indexed: 12/23/2022]
Abstract
AIMS/HYPOTHESIS Animal studies have indicated that disturbed diurnal rhythms of clock gene expression in adipose tissue can induce obesity and type 2 diabetes. The importance of the circadian timing system for energy metabolism is well established, but little is known about the diurnal regulation of (clock) gene expression in obese individuals with type 2 diabetes. In this study we aimed to identify key disturbances in the diurnal rhythms of the white adipose tissue transcriptome in obese individuals with type 2 diabetes. METHODS In a case-control design, we included six obese individuals with type 2 diabetes and six healthy, lean control individuals. All participants were provided with three identical meals per day for 3 days at zeitgeber time (ZT, with ZT 0:00 representing the time of lights on) 0:30, 6:00 and 11:30. Four sequential subcutaneous abdominal adipose tissue samples were obtained, on day 2 at ZT 15:30, and on day 3 at ZT 0:15, ZT 5:45 and ZT 11:15. Gene expression was measured using RNA sequencing. RESULTS The core clock genes showed reduced amplitude oscillations in the individuals with type 2 diabetes compared with the healthy control individuals. Moreover, in individuals with type 2 diabetes, only 1.8% (303 genes) of 16,818 expressed genes showed significant diurnal rhythmicity, compared with 8.4% (1421 genes) in healthy control individuals. Enrichment analysis revealed a loss of rhythm in individuals with type 2 diabetes of canonical metabolic pathways involved in the regulation of lipolysis. Enrichment analysis of genes with an altered mesor in individuals with type 2 diabetes showed decreased activity of the translation initiating pathway 'EIF2 signaling'. Individuals with type 2 diabetes showed a reduced diurnal rhythm in postprandial glucose concentrations. CONCLUSIONS/INTERPRETATION Diurnal clock and metabolic gene expression rhythms are decreased in subcutaneous adipose tissue of obese individuals with type 2 diabetes compared with lean control participants. Future investigation is needed to explore potential treatment targets as identified by our study, including clock enhancement and induction of EIF2 signalling. DATA AVAILABILITY The raw sequencing data and supplementary files for rhythmic expression analysis and Ingenuity Pathway Analysis have been deposited in NCBI Gene Expression Omnibus (GEO series accession number GSE104674).
Collapse
Affiliation(s)
- Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam UMC, location AMC, University of Amsterdam, room F5-162, P.O. Box 22660, 1100 DD, Amsterdam, the Netherlands.
| | - Aldo Jongejan
- Bioinformatics Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Sadaf Atiqi
- Department of Endocrinology and Metabolism, Amsterdam UMC, location AMC, University of Amsterdam, room F5-162, P.O. Box 22660, 1100 DD, Amsterdam, the Netherlands
| | - Jeroen P Vreijling
- Neurogenetics Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Eelkje J Limonard
- Department of Endocrinology and Metabolism, Amsterdam UMC, location AMC, University of Amsterdam, room F5-162, P.O. Box 22660, 1100 DD, Amsterdam, the Netherlands
| | - Erik Endert
- Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Frank Baas
- Neurogenetics Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Perry D Moerland
- Bioinformatics Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam UMC, location AMC, University of Amsterdam, room F5-162, P.O. Box 22660, 1100 DD, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, location AMC, University of Amsterdam, room F5-162, P.O. Box 22660, 1100 DD, Amsterdam, the Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam UMC, location AMC, University of Amsterdam, room F5-162, P.O. Box 22660, 1100 DD, Amsterdam, the Netherlands
| |
Collapse
|
28
|
Menko AS, Walker JL, Stepp MA. Fibrosis: Shared Lessons From the Lens and Cornea. Anat Rec (Hoboken) 2019; 303:1689-1702. [PMID: 30768772 PMCID: PMC6697240 DOI: 10.1002/ar.24088] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/23/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022]
Abstract
Regenerative repair in response to wounding involves cell proliferation and migration. This is followed by the reestablishment of cell structure and organization and a dynamic process of remodeling and restoration of the injured cells' extracellular matrix microenvironment and the integration of the newly synthesized matrix into the surrounding tissue. Fibrosis in the lungs, liver, and heart can lead to loss of life and in the eye to loss of vision. Learning to control fibrosis and restore normal tissue function after injury repair remains a goal of research in this area. Here we use knowledge gained using the lens and the cornea to provide insight into how fibrosis develops and clues to how it can be controlled. The lens and cornea are less complex than other tissues that develop life‐threatening fibrosis, but they are well characterized and research using them as model systems to study fibrosis is leading toward an improved understanding of fibrosis. Here we summarize the current state of the literature and how it is leading to promising new treatments. Anat Rec, 2019. © 2019 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- A Sue Menko
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Janice L Walker
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University, Washington, District of Columbia
| |
Collapse
|
29
|
Yang J, Song H, Chen L, Cao K, Zhang Y, Li Y, Hao X. Integrated analysis of microfibrillar-associated proteins reveals MFAP4 as a novel biomarker in human cancers. Epigenomics 2019; 11:1635-1651. [DOI: 10.2217/epi-2018-0080] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aim: The potential functions and underlying mechanism of microfibrillar-associated proteins (MFAPs) are explored in human cancers. Materials & methods: Here, we examined the expression profiles, prognostic values, epigenetic and genetic alterations of MFAPs in human cancers from public omics repository. Results: Among MFAPs family, MFAP4 was frequently downregulated in the most human cancers and high mRNA expression of MFAP4 significantly correlated with better overall survival in breast cancer. DNA hypermethylation in the promoter of MFAP4 decreased its mRNA expression. MFAP4 strongly associated with pathway in impairment and alteration of the elastic fibers. Conclusion: This integrated analysis provides new insights into MFAPs in human cancers and indicates that MFAP4 could be used as novel biomarker for developing therapies against human cancers.
Collapse
Affiliation(s)
- Jue Yang
- The State Key Laboratory of Functions & Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province & Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Hui Song
- The Key Laboratory of Endemic & Ethnic Diseases, Guizhou Medical University, Ministry of Education, Guiyang 550004, PR China
- The Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guizhou Province, Guiyang 550004, PR China
| | - Li Chen
- Guiyang University of Chinese Medicine, School of Pharmaceutical Sciences, Guiyang 550025, PR China
| | - Kun Cao
- Department of General Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550001, PR China
| | - Yongqiang Zhang
- Guizhou University, School of Pharmaceutical Sciences, Guiyang, 550025, PR China
| | - Yanmei Li
- The State Key Laboratory of Functions & Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province & Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Xiaojiang Hao
- The State Key Laboratory of Functions & Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province & Chinese Academic of Sciences, Guiyang 550014, PR China
| |
Collapse
|
30
|
Pahlavani M, Wijayatunga NN, Kalupahana NS, Ramalingam L, Gunaratne PH, Coarfa C, Rajapakshe K, Kottapalli P, Moustaid-Moussa N. Transcriptomic and microRNA analyses of gene networks regulated by eicosapentaenoic acid in brown adipose tissue of diet-induced obese mice. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1523-1531. [PMID: 30261280 PMCID: PMC6298436 DOI: 10.1016/j.bbalip.2018.09.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/01/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022]
Abstract
Brown adipose tissue (BAT) dissipates chemical energy as heat via thermogenesis and protects against obesity by increasing energy expenditure. However, regulation of BAT by dietary factors remains largely unexplored at the mechanistic level. We investigated the effect of eicosapentaenoic acid (EPA) on BAT metabolism. Male C57BL/6J (B6) mice were fed either a high-fat diet (HF, 45% kcal fat) or HF diet supplemented with EPA (HF-EPA, 6.75% kcal EPA) for 11 weeks. RNA sequencing (RNA-Seq) and microRNA (miRNA) profiling were performed on RNA from BAT using Illumina HiSeq and Illumina Genome Analyzer NextSeq, respectively. We conducted pathway analyses using ingenuity pathway analysis software (IPA®) and validated some genes and miRNAs using qPCR. We identified 479 genes that were differentially expressed (2-fold change, n = 3, P ≤ 0.05) in BAT from HF compared to HF-EPA. Genes negatively correlated with thermogenesis such as hypoxia inducible factor 1 alpha subunit inhibitor (Hif1an), were downregulated by EPA. Pathways related to thermogenesis such as peroxisome proliferator-activated receptor (PPAR) were upregulated by EPA while pathways associated with obesity and inflammation such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) were downregulated by EPA. MiRNA profiling identified nine and six miRNAs that were upregulated and downregulated by EPA, respectively (log2 fold change > 1.25, n = 3, P ≤ 0.05). Key regulatory miRNAs which were involved in thermogenesis, such as miR-455-3p and miR-129-5p were validated using qPCR. In conclusion, the depth of transcriptomic and miRNA profiling revealed novel mRNA-miRNA interaction networks in BAT which are involved in thermogenesis, and regulated by EPA.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States
| | - Nadeeja N Wijayatunga
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States
| | - Nishan S Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States; Department of Physiology, University of Peradeniya, Sri Lanka
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, United States
| | - Cristian Coarfa
- Department of Molecular and Cell Biology, Houston, TX, United States
| | - Kimal Rajapakshe
- Department of Molecular and Cell Biology, Houston, TX, United States
| | - Pratibha Kottapalli
- Center for Biotechnology and Genomics, Texas Tech University, Lubbock, TX, United States
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States.
| |
Collapse
|
31
|
Li Y, Wang X, Wang F, You L, Xu P, Cao Y, Chen L, Wen J, Guo X, Cui X, Ji C. Identification of intracellular peptides associated with thermogenesis in human brown adipocytes. J Cell Physiol 2018; 234:7104-7114. [PMID: 30387133 DOI: 10.1002/jcp.27465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Yun Li
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Xing Wang
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Fei Wang
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Lianghui You
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Pengfei Xu
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Yan Cao
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Ling Chen
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Juan Wen
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Xirong Guo
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Xianwei Cui
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| | - Chenbo Ji
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital) Nanjing China
| |
Collapse
|
32
|
Porter LC, Franczyk MP, Pietka T, Yamaguchi S, Lin JB, Sasaki Y, Verdin E, Apte RS, Yoshino J. NAD +-dependent deacetylase SIRT3 in adipocytes is dispensable for maintaining normal adipose tissue mitochondrial function and whole body metabolism. Am J Physiol Endocrinol Metab 2018; 315:E520-E530. [PMID: 29634313 PMCID: PMC6230701 DOI: 10.1152/ajpendo.00057.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mitochondrial dysfunction in adipose tissue is involved in the pathophysiology of obesity-induced systemic metabolic complications, such as type 2 diabetes, insulin resistance, and dyslipidemia. However, the mechanisms responsible for obesity-induced adipose tissue mitochondrial dysfunction are not clear. The aim of present study was to test the hypothesis that nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase sirtuin-3 (SIRT3) in adipocytes plays a critical role in adipose tissue mitochondrial biology and obesity. We first measured adipose tissue SIRT3 expression in obese and lean mice. Next, adipocyte-specific mitochondrial Sirt3 knockout (AMiSKO) mice were generated and metabolically characterized. We evaluated glucose and lipid metabolism in adult mice fed either a regular-chow diet or high-fat diet (HFD) and in aged mice. We also determined the effects of Sirt3 deletion on adipose tissue metabolism and mitochondrial biology. Supporting our hypothesis, obese mice had decreased SIRT3 gene and protein expression in adipose tissue. However, despite successful knockout of SIRT3, AMiSKO mice had normal glucose and lipid metabolism and did not change metabolic responses to HFD-feeding and aging. In addition, loss of SIRT3 had no major impact on putative SIRT3 targets, key metabolic pathways, and mitochondrial function in white and brown adipose tissue. Collectively, these findings suggest that adipocyte SIRT3 is dispensable for maintaining normal adipose tissue mitochondrial function and whole body metabolism. Contrary to our hypothesis, loss of SIRT3 function in adipocytes is unlikely to contribute to the pathophysiology of obesity-induced metabolic complications.
Collapse
Affiliation(s)
- Lane C Porter
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine , St. Louis, Missouri
| | - Michael P Franczyk
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine , St. Louis, Missouri
| | - Terri Pietka
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine , St. Louis, Missouri
| | - Shintaro Yamaguchi
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine , St. Louis, Missouri
| | - Jonathan B Lin
- Department of Ophthalmology, Washington University School of Medicine , St. Louis, Missouri
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine , St. Louis, Missouri
| | - Eric Verdin
- Gladstone Institutes, University of California San Francisco , San Francisco, California
- Buck Institute for Research on Aging , Novato, California
| | - Rajendra S Apte
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine , St. Louis, Missouri
- Department of Ophthalmology, Washington University School of Medicine , St. Louis, Missouri
- Department of Developmental Biology, Washington University School of Medicine , St. Louis, Missouri
| | - Jun Yoshino
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine , St. Louis, Missouri
| |
Collapse
|
33
|
Trivanović D, Drvenica I, Kukolj T, Obradović H, Okić Djordjević I, Mojsilović S, Krstić J, Bugarski B, Jauković A, Bugarski D. Adipoinductive effect of extracellular matrix involves cytoskeleton changes and SIRT1 activity in adipose tissue stem/stromal cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S370-S382. [PMID: 30198336 DOI: 10.1080/21691401.2018.1494183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Adipose tissue (AT) homeostasis and expansion are dependent on complex crosstalk between resident adipose stromal/stem cells (ASCs) and AT extracellular matrix (ECM). Although adipose tissue ECM (atECM) is one of the key players in the stem cell niche, data on bidirectional interaction of ASCs and atECM are still scarce. Here, we investigated how atECM guides ASCs' differentiation. atECM altered shape and cytoskeleton organization of ASCs without changing their proliferation, β-galactosidase activity and adhesion. Cytoskeleton modifications occurred due to fostered parallel organization of F-actin and elevated expression of Vimentin in ASCs. After seven-day cultivation, atECM impaired osteogenesis of ASCs, simultaneously decreasing expression of Runx2. In addition, atECM accelerated early adipogenesis concomitantly with altered Vimentin organization in ASCs, slightly increasing PPARγ, while elevated Adiponectin and Vimentin mRNA expression. Early adipogenesis triggered by atECM was followed by upregulated mitochondrial activity and Sirtuin 1 (SIRT1) expression in ASCs. Proadipogenic events induced by atECM were mediated by SIRT1, indicating the supportive role of atECM in adipogenesis-related metabolic state of ASCs. These results provide a closer look at the effects of atECM on ASC physiology and may support the advancement of engineering design in soft tissue reconstruction and fundamental research of AT.
Collapse
Affiliation(s)
- Drenka Trivanović
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Ivana Drvenica
- b Laboratory for Immunology , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Tamara Kukolj
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Hristina Obradović
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Ivana Okić Djordjević
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Slavko Mojsilović
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Jelena Krstić
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Branko Bugarski
- c Department of Chemical Engineering, Faculty of Technology and Metallurgy , University of Belgrade , Belgrade , Serbia
| | - Aleksandra Jauković
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Diana Bugarski
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| |
Collapse
|
34
|
Romier B, Ivaldi C, Sartelet H, Heinz A, Schmelzer CEH, Garnotel R, Guillot A, Jonquet J, Bertin E, Guéant JL, Alberto JM, Bronowicki JP, Amoyel J, Hocine T, Duca L, Maurice P, Bennasroune A, Martiny L, Debelle L, Durlach V, Blaise S. Production of Elastin-Derived Peptides Contributes to the Development of Nonalcoholic Steatohepatitis. Diabetes 2018; 67:1604-1615. [PMID: 29802129 DOI: 10.2337/db17-0490] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/14/2018] [Indexed: 11/13/2022]
Abstract
Affecting more than 30% of the Western population, nonalcoholic fatty liver disease (NAFLD) is the most common liver disease and can lead to multiple complications, including nonalcoholic steatohepatitis (NASH), cancer, hypertension, and atherosclerosis. Insulin resistance and obesity are described as potential causes of NAFLD. However, we surmised that factors such as extracellular matrix remodeling of large blood vessels, skin, or lungs may also participate in the progression of liver diseases. We studied the effects of elastin-derived peptides (EDPs), biomarkers of aging, on NAFLD progression. We evaluated the consequences of EDP accumulation in mice and of elastin receptor complex (ERC) activation on lipid storage in hepatocytes, inflammation, and fibrosis development. The accumulation of EDPs induces hepatic lipogenesis (i.e., SREBP1c and ACC), inflammation (i.e., Kupffer cells, IL-1β, and TGF-β), and fibrosis (collagen and elastin expression). These effects are induced by inhibition of the LKB1-AMPK pathway by ERC activation. In addition, pharmacological inhibitors of EDPs demonstrate that this EDP-driven lipogenesis and fibrosis relies on engagement of the ERC. Our data reveal a major role of EDPs in the development of NASH, and they provide new clues for understanding the relationship between NAFLD and vascular aging.
Collapse
Affiliation(s)
- Béatrice Romier
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Corinne Ivaldi
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Hervé Sartelet
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Andrea Heinz
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Christian E H Schmelzer
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany
| | - Roselyne Garnotel
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Alexandre Guillot
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Jessica Jonquet
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Eric Bertin
- Champagne Ardenne Specialized Center in Obesity, University Hospital Center, Reims, France
| | - Jean-Louis Guéant
- Institut National de la Santé et de la Recherche Médicale, U954, and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France
- Department of Molecular Medicine and Personalized Therapeutics and Department of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France
| | - Jean-Marc Alberto
- Institut National de la Santé et de la Recherche Médicale, U954, and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France
| | - Jean-Pierre Bronowicki
- Institut National de la Santé et de la Recherche Médicale, U954, and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France
- Department of Gastroenterology and Hepatology, University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France
| | - Johanne Amoyel
- Department of Gastroenterology and Hepatology, University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France
| | - Thinhinane Hocine
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Amar Bennasroune
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Laurent Martiny
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Laurent Debelle
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Vincent Durlach
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | - Sébastien Blaise
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
35
|
Wang JK, Wang WJ, Cai HY, Du BB, Mai P, Zhang LJ, Ma W, Hu YG, Feng SF, Miao GY. MFAP2 promotes epithelial-mesenchymal transition in gastric cancer cells by activating TGF-β/SMAD2/3 signaling pathway. Onco Targets Ther 2018; 11:4001-4017. [PMID: 30034240 PMCID: PMC6047603 DOI: 10.2147/ott.s160831] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Introduction Microfibril-associated protein 2 (MFAP2) is an extracellular matrix protein that interacts with fibrillin to modulate the function of microfibrils. MFAP2 has been reported to play a significant role in obesity, diabetes, and osteopenia, and has been shown to be upregulated in head and neck squamous cell carcinoma. However, the molecular function and prognostic value of MFAP2 have never been reported in gastric cancer (GC) or any other tumors. Methods The current study investigated the expression patterns, prognostic significance, functional role, and possible mechanisms of MFAP2 in GC. Results We demonstrated that MFAP2 was overexpressed in GC tissues, and its overexpression was significantly correlated with poor overall and disease-free survival in patients with GC. Moreover, we found that MFAP2 promoted the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) phenotype in GC cells. MFAP2 might modulate EMT of GC cells by activating the TGF-β/SMAD2/3 signaling pathway. Conclusion These findings provide novel evidence that MFAP2 plays a crucial role in the progression of GC. Therefore, MFAP2 may be a promising prognostic marker and a potent anticancer agent.
Collapse
Affiliation(s)
- Jian-Kai Wang
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Wen-Juan Wang
- Physical Examination Center, The Third People's Hospital of Gansu, Lanzhou, Gansu 730000, China
| | - Hong-Yi Cai
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Bin-Bin Du
- Department of Anorectal Surgery, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Ping Mai
- Department of Gastroenterology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Li-Juan Zhang
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Wen Ma
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Yong-Guo Hu
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Shi-Fang Feng
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Guo-Ying Miao
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| |
Collapse
|
36
|
Turecamo SE, Walji TA, Broekelmann TJ, Williams JW, Ivanov S, Wee NK, Procknow JD, McManus MR, Randolph GJ, Scheller EL, Mecham RP, Craft CS. Contribution of metabolic disease to bone fragility in MAGP1-deficient mice. Matrix Biol 2018; 67:1-14. [PMID: 29519758 PMCID: PMC6077988 DOI: 10.1016/j.matbio.2018.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 12/16/2022]
Abstract
Microfibril-associated glycoprotein-1 (MAGP1) is an extracellular matrix protein that interacts with fibrillin and is involved in regulating the bioavailability of signaling molecules such as TGFβ. Mice with germline MAGP1 deficiency (Mfap2-/-) develop increased adiposity, hyperglycemia, insulin resistance, bone marrow adipose tissue expansion, reduced cancellous bone mass, cortical bone thinning and bone fragility. The goal of this study was to assess whether the Mfap2-/- bone phenotypes were due to loss of MAGP1 locally or secondary to a change in whole body physiology (metabolic dysfunction). To do this, mice with conditional deletion of MAGP1 in the limb skeleton were generated by crossing MAGP1-flox mice (Mfap2lox/lox) with Prx1-Cre mice. Mfap2Prx-/- mice did not show any changes in peripheral adiposity, hyperglycemia or insulin sensitivity, but did have increased bone length and cancellous bone loss that was comparable to the germline Mfap2-/- knockout. Unlike the germline knockout, marrow adiposity, cortical bone thickness and bone strength in Mfap2Prx-/- mice were normal. These findings implicate systemic metabolic dysfunction in the development of bone fragility in germline Mfap2-/- mice. An unexpected finding of this study was the detection of MAGP1 protein in the Mfap2Prx-/- hematopoietic bone marrow, despite the absence of MAGP1 protein in osseous bone matrix and absent Mfap2 transcript expression at both sites. This suggests MAGP1 from a secondary site may accumulate in the bone marrow, but not be incorporated into the bone matrix, during times of regional MAGP1 depletion.
Collapse
Affiliation(s)
- S E Turecamo
- Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - T A Walji
- Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - T J Broekelmann
- Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - J W Williams
- Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - S Ivanov
- INSERM U1065, Mediterranean Center of Molecular Medicine, University of Nice Sophia-Antipolis, Faculty of Medicine, Nice, France.
| | - N K Wee
- Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - J D Procknow
- Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - M R McManus
- Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - G J Randolph
- Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - E L Scheller
- Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA; Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - R P Mecham
- Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - C S Craft
- Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA; Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
37
|
Abstract
Microfibril-associated glycoproteins 1 and 2 (MAGP-1, MAGP-2) are protein components of extracellular matrix microfibrils. These proteins interact with fibrillin, the core component of microfibrils, and impart unique biological properties that influence microfibril function in vertebrates. MAGPs bind active forms of TGFβ and BMPs and are capable of modulating Notch signaling. Mutations in MAGP-1 or MAGP-2 have been linked to thoracic aneurysms and metabolic disease in humans. MAGP-2 has also been shown to be an important biomarker in several human cancers. Mice lacking MAGP-1 or MAGP-2 have defects in multiple organ systems, which reflects the widespread distribution of microfibrils in vertebrate tissues. This review summarizes our current understanding of the function of the MAGPs and their relationship to human disease.
Collapse
Affiliation(s)
- Clarissa S Craft
- Division of Bone and Mineral Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Thomas J Broekelmann
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, United States.
| |
Collapse
|
38
|
Abstract
Brown adipose tissue aging and the concomitant loss of thermogenic capacity have been linked to an inability to maintain normal energy homeostasis in late life. Similarly, the ability of white fat to convert into brite/beige adipose tissue declines. This may ultimately exacerbate the progression of age-related metabolic pathologies, such as insulin resistance and obesity. The depletion of all types of brown adipocytes during aging is well-established and has been described in rodent models as well as humans. We here review the available literature on the potential mechanisms leading to cell-autonomous and microenvironment-related aspects of brown adipocyte dysfunction. Among these, cellular senescence, mitochondrial impairment, and deteriorating changes to the local and endocrine microenvironments have been proposed. An important goal of aging research is to develop approaches that may not only extend life expectancy but also prolong health-span. These efforts may also be aimed at maintaining metabolic health throughout life by targeting brown adipocyte function.
Collapse
Affiliation(s)
- Antonia Graja
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany. .,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany. .,German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany.
| |
Collapse
|
39
|
Lark DS, Wasserman DH. Meta-fibrosis links positive energy balance and mitochondrial metabolism to insulin resistance. F1000Res 2017; 6:1758. [PMID: 29043068 PMCID: PMC5621108 DOI: 10.12688/f1000research.11653.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2017] [Indexed: 12/12/2022] Open
Abstract
Obesity and insulin resistance often emerge from positive energy balance and generally are linked to low-grade inflammation. This low-grade inflammation has been called “meta-inflammation” because it is a consequence of the metabolic dysregulation that can accompany overnutrition. One means by which meta-inflammation is linked to insulin resistance is extracellular matrix expansion secondary to meta-inflammation, which we define here as “meta-fibrosis”. The significance of meta-fibrosis is that it reflects a situation in which the extracellular matrix functions as a multi-level integrator of local (for example, mitochondrial reactive oxygen species production) and systemic (for example, inflammation) inputs that couple to cellular processes creating insulin resistance. While adipose tissue extracellular matrix remodeling has received considerable attention, it is becoming increasingly apparent that liver and skeletal muscle extracellular matrix remodeling also contributes to insulin resistance. In this review, we address recent advances in our understanding of energy balance, mitochondrial energetics, meta-inflammation, and meta-fibrosis in the development of insulin resistance.
Collapse
Affiliation(s)
- Daniel S Lark
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
40
|
Adipose Tissue Function and Expandability as Determinants of Lipotoxicity and the Metabolic Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:161-196. [PMID: 28585199 DOI: 10.1007/978-3-319-48382-5_7] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis and it is constituted of three different types of adipocytes : white, beige and brown which are integrated with vascular, immune, neural and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concert action of the three type of adipocytes/tissues has been reported to ensure an optimal metabolic status in rodents. However, when one or multiple of these adipose depots become dysfunctional as a consequence of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity counteracts obesity and its associated lipotoxic metabolic effects. The development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition and expandability capacity as well as molecular and metabolic signatures in both physiological and pathophysiological conditions.
Collapse
|
41
|
Pervin S, Singh V, Tucker A, Collazo J, Singh R. Modulation of transforming growth factor-β/follistatin signaling and white adipose browning: therapeutic implications for obesity related disorders. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0036/hmbci-2017-0036.xml. [PMID: 28888087 DOI: 10.1515/hmbci-2017-0036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/17/2017] [Indexed: 12/15/2022]
Abstract
Obesity is a major risk factor for the development of diabetes, insulin resistance, dyslipidemia, cardiovascular disease and other related metabolic conditions. Obesity develops from perturbations in overall cellular bioenergetics when energy intake chronically exceeds total energy expenditure. Lifestyle interventions based on reducing total energy uptake and increasing activities including exercise have proved ineffective in the prevention and treatment of obesity because of poor adherence to such interventions for an extended period of time. Brown adipose tissue (BAT) has an extraordinary metabolic capacity to burn excess stored energy and holds great promise in combating obesity and related diseases. This unique ability to nullify the effects of extra energy intake of these specialized tissues has provided attractive perspectives for the therapeutic potential of BAT in humans. Browning of white adipose tissue by promoting the expression and activity of key mitochondrial uncoupling protein 1 (UCP1) represents an exciting new strategy to combat obesity via enhanced energy dissipation. Members of the transforming growth factor-beta (TGF-β) superfamily including myostatin and follistatin have recently been demonstrated to play a key role in regulating white adipose browning both in in-vitro and in-vivo animal models and thereby present attractive avenues for exploring the therapeutic potential for the treatment of obesity and related metabolic diseases.
Collapse
|
42
|
Nawaz A, Aminuddin A, Kado T, Takikawa A, Yamamoto S, Tsuneyama K, Igarashi Y, Ikutani M, Nishida Y, Nagai Y, Takatsu K, Imura J, Sasahara M, Okazaki Y, Ueki K, Okamura T, Tokuyama K, Ando A, Matsumoto M, Mori H, Nakagawa T, Kobayashi N, Saeki K, Usui I, Fujisaka S, Tobe K. CD206 + M2-like macrophages regulate systemic glucose metabolism by inhibiting proliferation of adipocyte progenitors. Nat Commun 2017; 8:286. [PMID: 28819169 PMCID: PMC5561263 DOI: 10.1038/s41467-017-00231-1] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 06/08/2017] [Indexed: 01/06/2023] Open
Abstract
Adipose tissue resident macrophages have important roles in the maintenance of tissue homeostasis and regulate insulin sensitivity for example by secreting pro-inflammatory or anti-inflammatory cytokines. Here, we show that M2-like macrophages in adipose tissue regulate systemic glucose homeostasis by inhibiting adipocyte progenitor proliferation via the CD206/TGFβ signaling pathway. We show that adipose tissue CD206+ cells are primarily M2-like macrophages, and ablation of CD206+ M2-like macrophages improves systemic insulin sensitivity, which was associated with an increased number of smaller adipocytes. Mice genetically engineered to have reduced numbers of CD206+ M2-like macrophages show a down-regulation of TGFβ signaling in adipose tissue, together with up-regulated proliferation and differentiation of adipocyte progenitors. Our findings indicate that CD206+ M2-like macrophages in adipose tissues create a microenvironment that inhibits growth and differentiation of adipocyte progenitors and, thereby, control adiposity and systemic insulin sensitivity.Adipose tissue contains macrophages that can influence both local and systemic metabolism via the secretion of cytokines. Here, Nawaz et al. report that M2-like macrophages, present in adipose tissue, create a microenvironment that inhibits proliferation of adipocyte progenitors due to the secretion of TGF-β1.
Collapse
Affiliation(s)
- Allah Nawaz
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Aminuddin Aminuddin
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.,Department of Nutrition, Faculty of Medicine, University of Hasanuddin, Makassar, Kota Makassar, Sulawesi Selatan, 90245, Indonesia
| | - Tomonobu Kado
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Akiko Takikawa
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Seiji Yamamoto
- Department of Pathology, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Koichi Tsuneyama
- Department of Diagnostic Pathology, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.,Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Yoshiko Igarashi
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Masashi Ikutani
- Department of Immune Regulation, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Yasuhiro Nishida
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.,JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Kiyoshi Takatsu
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.,Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Imiz-shi, Toyama, 939-0363, Japan
| | - Johji Imura
- Department of Diagnostic Pathology, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Masakiyo Sasahara
- Department of Pathology, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Yukiko Okazaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Kohjiro Ueki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.,Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan.,Section of Animal Models, Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Kumpei Tokuyama
- Doctoral Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8574, Japan
| | - Akira Ando
- Doctoral Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8574, Japan
| | - Michihiro Matsumoto
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Takashi Nakagawa
- Department of Metabolism and Nutrition, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Norihiko Kobayashi
- Department of Disease Control, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Kumiko Saeki
- Department of Disease Control, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Isao Usui
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.
| |
Collapse
|
43
|
Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Invest 2017; 127:74-82. [PMID: 28045400 DOI: 10.1172/jci88883] [Citation(s) in RCA: 503] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
There are three dominant contributors to the pathogenesis of dysfunctional adipose tissue (AT) in obesity: unresolved inflammation, inappropriate extracellular matrix (ECM) remodeling and insufficient angiogenic potential. The interactions of these processes during AT expansion reflect both a linear progression as well as feed-forward mechanisms. For example, both inflammation and inadequate angiogenic remodeling can drive fibrosis, which can in turn promote migration of immune cells into adipose depots and impede further angiogenesis. Therefore, the relationship between the members of this triad is complex but important for our understanding of the pathogenesis of obesity. Here we untangle some of these intricacies to highlight the contributions of inflammation, angiogenesis, and the ECM to both "healthy" and "unhealthy" AT expansion.
Collapse
|
44
|
Walji TA, Turecamo SE, DeMarsilis AJ, Sakai LY, Mecham RP, Craft CS. Characterization of metabolic health in mouse models of fibrillin-1 perturbation. Matrix Biol 2016; 55:63-76. [PMID: 26902431 PMCID: PMC4992667 DOI: 10.1016/j.matbio.2016.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/05/2016] [Accepted: 02/05/2016] [Indexed: 11/16/2022]
Abstract
Mutations in the microfibrillar protein fibrillin-1 or the absence of its binding partner microfibril-associated glycoprotein (MAGP1) lead to increased TGFβ signaling due to an inability to sequester latent or active forms of TGFβ, respectively. Mouse models of excess TGFβ signaling display increased adiposity and predisposition to type-2 diabetes. It is therefore interesting that individuals with Marfan syndrome, a disease in which fibrillin-1 mutation leads to aberrant TGFβ signaling, typically present with extreme fat hypoplasia. The goal of this project was to characterize multiple fibrillin-1 mutant mouse strains to understand how fibrillin-1 contributes to metabolic health. The results of this study demonstrate that fibrillin-1 contributes little to lipid storage and metabolic homeostasis, which is in contrast to the obesity and metabolic changes associated with MAGP1 deficiency. MAGP1 but not fibrillin-1 mutant mice had elevated TGFβ signaling in their adipose tissue, which is consistent with the difference in obesity phenotypes. However, fibrillin-1 mutant strains and MAGP1-deficient mice all exhibit increased bone length and reduced bone mineralization which are characteristic of Marfan syndrome. Our findings suggest that Marfan-associated adipocyte hypoplasia is likely not due to microfibril-associated changes in adipose tissue, and provide evidence that MAGP1 may function independently of fibrillin in some tissues.
Collapse
Affiliation(s)
- Tezin A Walji
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sarah E Turecamo
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Antea J DeMarsilis
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lynn Y Sakai
- Department of Biochemistry & Molecular Biology, Molecular & Medical Genetics, Oregon Health & Science University, Shriners Hospital for Children, Portland, OR 97201, USA
| | - Robert P Mecham
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Clarissa S Craft
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
45
|
Pellegrinelli V, Carobbio S, Vidal-Puig A. Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues. Diabetologia 2016; 59:1075-88. [PMID: 27039901 PMCID: PMC4861754 DOI: 10.1007/s00125-016-3933-4] [Citation(s) in RCA: 278] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/23/2016] [Indexed: 02/07/2023]
Abstract
White adipose tissue (WAT) has key metabolic and endocrine functions and plays a role in regulating energy homeostasis and insulin sensitivity. WAT is characterised by its capacity to adapt and expand in response to surplus energy through processes of adipocyte hypertrophy and/or recruitment and proliferation of precursor cells in combination with vascular and extracellular matrix remodelling. However, in the context of sustained obesity, WAT undergoes fibro-inflammation, which compromises its functionality, contributing to increased risk of type 2 diabetes and cardiovascular diseases. Conversely, brown adipose tissue (BAT) and browning of WAT represent potential therapeutic approaches, since dysfunctional white adipocyte-induced lipid overspill can be halted by BAT/browning-mediated oxidative anti-lipotoxic effects. Better understanding of the cellular and molecular pathophysiological mechanisms regulating adipocyte size, number and depot-dependent expansion has become a focus of interest over recent decades. Here, we summarise the mechanisms contributing to adipose tissue (AT) plasticity and function including characteristics and cellular complexity of the various adipose depots and we discuss recent insights into AT origins, identification of adipose precursors, pathophysiological regulation of adipogenesis and its relation to WAT/BAT expandability in obesity and its associated comorbidities.
Collapse
Affiliation(s)
- Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK.
| | - Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK.
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK.
| |
Collapse
|
46
|
Walji TA, Turecamo SE, Sanchez AC, Anthony BA, Abou-Ezzi G, Scheller EL, Link DC, Mecham RP, Craft CS. Marrow Adipose Tissue Expansion Coincides with Insulin Resistance in MAGP1-Deficient Mice. Front Endocrinol (Lausanne) 2016; 7:87. [PMID: 27445989 PMCID: PMC4928449 DOI: 10.3389/fendo.2016.00087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/22/2016] [Indexed: 12/21/2022] Open
Abstract
Marrow adipose tissue (MAT) is an endocrine organ with the potential to influence skeletal remodeling and hematopoiesis. Pathologic MAT expansion has been studied in the context of severe metabolic challenge, including caloric restriction, high fat diet feeding, and leptin deficiency. However, the rapid change in peripheral fat and glucose metabolism associated with these models impedes our ability to examine which metabolic parameters precede or coincide with MAT expansion. Microfibril-associated glycoprotein-1 (MAGP1) is a matricellular protein that influences cellular processes by tethering signaling molecules to extracellular matrix structures. MAGP1-deficient (Mfap2 (-/-)) mice display a progressive excess adiposity phenotype, which precedes insulin resistance and occurs without changes in caloric intake or ambulation. Mfap2 (-/-) mice were, therefore, used as a model to associate parameters of metabolic disease, bone remodeling, and hematopoiesis with MAT expansion. Marrow adiposity was normal in Mfap2 (-/-) mice until 6 months of age; however, by 10 months, marrow fat volume had increased fivefold relative to wild-type control at the same age. Increased gonadal fat pad mass and hyperglycemia were detectable in Mfap2 (-/-) mice by 2 months, but peaked by 6 months. The development of insulin resistance coincided with MAT expansion. Longitudinal characterization of bone mass demonstrated a disconnection in MAT volume and bone volume. Specifically, Mfap2 (-/-) mice had reduced trabecular bone volume by 2 months, but this phenotype did not progress with age or MAT expansion. Interestingly, MAT expansion in the 10-month-old Mfap2 (-/-) mice was associated with modest alterations in basal hematopoiesis, including a shift from granulopoiesis to B lymphopoiesis. Together, these findings indicate MAT expansion is coincident with insulin resistance, but not excess peripheral adiposity or hyperglycemia in Mfap2 (-/-) mice; and substantial MAT accumulation does not necessitate a proportional decrease in either bone mass or bone marrow cellularity.
Collapse
Affiliation(s)
- Tezin A. Walji
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah E. Turecamo
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alejandro Coca Sanchez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcala de Henares, Madrid, Spain
| | - Bryan A. Anthony
- Department of Medicine, Oncology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Grazia Abou-Ezzi
- Department of Medicine, Oncology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Erica L. Scheller
- Department of Medicine, Bone and Mineral Diseases Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel C. Link
- Department of Medicine, Oncology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert P. Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Clarissa S. Craft
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Bone and Mineral Diseases Division, Washington University School of Medicine, St. Louis, MO, USA
- *Correspondence: Clarissa S. Craft,
| |
Collapse
|
47
|
Vassequi-Silva T, Pereira DS, Nery Diez ACC, Braga GG, Godoy JA, Mendes CB, Dos Santos L, Krieger JE, Antunes E, Costa FTM, Vicente CP, Werneck CC. Losartan and captopril treatment rescue normal thrombus formation in microfibril associated glycoprotein-1 (MAGP1) deficient mice. Thromb Res 2015; 138:7-15. [PMID: 26826502 DOI: 10.1016/j.thromres.2015.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 11/10/2015] [Accepted: 12/09/2015] [Indexed: 11/18/2022]
Abstract
INTRODUCTION MAGP1 is a glycoprotein present in the elastic fibers and is a part of the microfibrils components. MAGP1 interacts with von Willebrand factor and the active form of TGF-β and BMP. In mice lacking MAGP1, thrombus formation is delayed, increasing the occlusion time of carotid artery despite presenting normal blood coagulation in vitro. MAGP1-containing microfibrils may play a role in hemostasis and thrombosis. In this work, we evaluated the function of MAGP1 and its relation to TGF-β in the arterial thrombosis process. METHODS AND RESULTS We analyzed thrombus formation time in wild type and MAGP1-deficient mice comparing Rose Bengal and Ferric Chloride induced arterial lesion. The potential participation of TGF-β in this process was accessed when we treated both wild type and MAGP1-deficient mice with losartan (an antihypertensive drug that decreases TGF-β activity) or captopril (an angiotensin converting enzyme inhibitor that was used as a control antihypertensive drug). Besides, we evaluated thrombus embolization and the gelatinolytic activity in the arterial walls in vitro and ex vivo. Losartan and captopril were able to recover the thrombus formation time without changing blood pressure, activated partial thromboplastin time (aPTT), PT (prothrombin time), platelet aggregation and adhesion, but decreased gelatinase activity. CONCLUSIONS Our results suggest that both treatments are effective in the prevention of the sub-endothelial ECM degradation, allowing the recovery of normal thrombus formation.
Collapse
Affiliation(s)
| | | | | | - Guilherme G Braga
- Department of Biochemistry and Tissue Biology, State University of Campinas, SP, Brazil
| | - Juliana A Godoy
- Department of Structural and Functional Biology, State University of Campinas, SP, Brazil
| | - Camila B Mendes
- Department of Pharmacology, State University of Campinas, SP, Brazil
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espirito Santo, ES, Brazil
| | - José E Krieger
- Laboratory of Genetic and Molecular Cardiology, InCor-HC/FMUSP, SP, Brazil
| | - Edson Antunes
- Department of Pharmacology, State University of Campinas, SP, Brazil
| | - Fábio T M Costa
- Department of Genetics and Evolution and Bioagents, State University of Campinas, SP, Brazil
| | - Cristina P Vicente
- Department of Structural and Functional Biology, State University of Campinas, SP, Brazil
| | - Claudio C Werneck
- Department of Biochemistry and Tissue Biology, State University of Campinas, SP, Brazil.
| |
Collapse
|
48
|
Vaittinen M, Kolehmainen M, Rydén M, Eskelinen M, Wabitsch M, Pihlajamäki J, Uusitupa M, Pulkkinen L. MFAP5 is related to obesity-associated adipose tissue and extracellular matrix remodeling and inflammation. Obesity (Silver Spring) 2015; 23:1371-8. [PMID: 26054006 DOI: 10.1002/oby.21103] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/03/2015] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Low-grade inflammation is involved in adipose tissue (AT) and extracellular matrix (ECM) remodeling and induces deposition of ECM proteins in AT. We have previously shown that MFAP5 (microfibrillar-associated protein 5) expression decreases in AT after weight loss. The aim of this study was to investigate MFAP5 localization in human AT and gene expression in adipocytes and the role of MFAP5 in adipocyte metabolism and inflammation. METHODS MFAP5 protein localization and gene expression were studied with immunohistochemistry and quantitative reverse transcriptase PCR (RT-qPCR) in human subcutaneous AT and cultured Simpson-Golabi-Behmel syndrome (SGBS) adipocytes, respectively. The effect of MFAP5 knock-down by siRNA on gene expression and insulin action was examined with RT-qPCR, western blot, and insulin-stimulated glucose uptake. The effect of different cytokines on MFAP5 gene and protein expression was investigated in cultured human SGBS preadipocytes. RESULTS MFAP5 protein was highly expressed in AT, and gene expression decreased during adipocyte differentiation in SGBS cells. Treatment of preadipocytes with TNFα and TGFβ1 increased MFAP5 gene and protein expression. Furthermore, MFAP5 knock-down decreased the expression of genes involved in inflammation. CONCLUSIONS Our results demonstrate that factors involving low-grade inflammation modulate MFAP5 expression and that the modified expression of MFAP5 may further regulate AT inflammation.
Collapse
Affiliation(s)
- Maija Vaittinen
- Department of Clinical Nutrition, University of Eastern Finland, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Marjukka Kolehmainen
- Department of Clinical Nutrition, University of Eastern Finland, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institute, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Matti Eskelinen
- Department of Surgery, Kuopio University Hospital, Kuopio, Finland
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Jussi Pihlajamäki
- Department of Clinical Nutrition, University of Eastern Finland, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
- Departments of Medicine and Clinical Nutrition, Obesity Center, Kuopio University Hospital, Kuopio, Finland
| | - Matti Uusitupa
- Department of Clinical Nutrition, University of Eastern Finland, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
- Research Unit, Kuopio University Hospital, Kuopio, Finland
| | - Leena Pulkkinen
- Department of Clinical Nutrition, University of Eastern Finland, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| |
Collapse
|
49
|
Mecham RP, Gibson MA. The microfibril-associated glycoproteins (MAGPs) and the microfibrillar niche. Matrix Biol 2015; 47:13-33. [PMID: 25963142 DOI: 10.1016/j.matbio.2015.05.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 10/23/2022]
Abstract
The microfibril-associated glycoproteins MAGP-1 and MAGP-2 are extracellular matrix proteins that interact with fibrillin to influence microfibril function. The two proteins are related through a 60 amino acid matrix-binding domain but their sequences differ outside of this region. A distinguishing feature of both proteins is their ability to interact with TGFβ family growth factors, Notch and Notch ligands, and multiple elastic fiber proteins. MAGP-2 can also interact with αvβ3 integrins via a RGD sequence that is not found in MAGP-1. Morpholino knockdown of MAGP-1 expression in zebrafish resulted in abnormal vessel wall architecture and altered vascular network formation. In the mouse, MAGP-1 deficiency had little effect on elastic fibers in blood vessels and lung but resulted in numerous unexpected phenotypes including bone abnormalities, hematopoietic changes, increased fat deposition, diabetes, impaired wound repair, and a bleeding diathesis. Inactivation of the gene for MAGP-2 in mice produced a neutropenia yet had minimal effects on bone or adipose homeostasis. Double knockouts had phenotypes characteristic of each individual knockout as well as several additional traits only seen when both genes are inactivated. A common mechanism underlying all of the traits associated with the knockout phenotypes is altered TGFβ signaling. This review summarizes our current understanding of the function of the MAGPs and discusses ideas related to their role in growth factor regulation.
Collapse
Affiliation(s)
- Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Mark A Gibson
- School of Medical Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
50
|
Craft CS. MAGP1, the extracellular matrix, and metabolism. Adipocyte 2015; 4:60-4. [PMID: 26167404 DOI: 10.4161/adip.32209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/23/2014] [Accepted: 07/29/2014] [Indexed: 01/11/2023] Open
Abstract
Adipose tissue and the extracellular matrix were once considered passive players in regulating physiological processes. Now, both entities are acknowledged for their capacity to engage signal transduction pathways, and for their involvement in maintaining normal tissue homeostasis. We recently published a series of studies that identified a novel mechanism whereby an extracellular matrix molecule, MAGP1 (microfibril associated glycoprotein 1), can regulate energy metabolism in adipose tissue. MAGP1 is a component of extracellular microfibrils and plays a supportive role in maintaining thermoregulation by indirectly regulating expression of the thermogenic uncoupling proteins (UCPs). The focus of this commentary is to draw attention to the role of the extracellular matrix in regulating the bioavailability of signaling molecules, like transforming growth factor β (TGFβ), and exemplify that a better understanding of the extracellular matrix's biological properties could unveil a new source of therapeutic targets for metabolic diseases.
Collapse
|