1
|
Yamada K, Kubota Y, Kosaka K, Yamaji Y, Akita S, Kuroda M, Mitsukawa N. Emergence of regulatory and matrix-producing stromal cells during adipogenic induction in human ceiling culture-derived preadipocytes. Biochem Biophys Res Commun 2025; 771:152012. [PMID: 40409116 DOI: 10.1016/j.bbrc.2025.152012] [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: 05/04/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025]
Abstract
Ceiling culture-derived preadipocytes (ccdPAs) are fibroblast-like cells believed to originate from mature adipocytes after the loss of lipid droplets. Unlike adipose-derived stem cells (ASCs), which are heterogeneous, ccdPAs are considered a more homogeneous population. However, their response to adipogenic differentiation stimuli at the single-cell level has not been fully characterized. In this study, we performed single-cell RNA sequencing (scRNA-seq) and epigenetic analyses to investigate early transcriptomic changes following adipogenic induction in ASCs and ccdPAs. scRNA-seq revealed that, in addition to preadipocytes/adipocytes, a substantial population of regulatory or structural cells, characterized by the expression of genes associated with extracellular matrix organization, structural support, and cell-cell interactions, such as F3 and MGP, emerged in both ASCs and ccdPAs. Notably, ATOH8, a transcription factor with limited prior characterization in adipogenesis, showed markedly higher expression in ccdPAs than in ASCs. This was supported by epigenetic analyses demonstrating lower CpG methylation and higher H3K4me3 levels at the ATOH8 locus in ccdPAs. Our findings suggest that adipogenic induction of ccdPAs generates diverse cell populations. This study provides the first single-cell level insight into the adipogenic response of primary cultured human ccdPAs.
Collapse
Affiliation(s)
- Kahoko Yamada
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Yoshitaka Kubota
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan.
| | - Kentaro Kosaka
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Yoshihisa Yamaji
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Shinsuke Akita
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Masayuki Kuroda
- Center for Advanced Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Nobuyuki Mitsukawa
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| |
Collapse
|
2
|
Zhang WZ, Wu CY, Lai H. A Review on the Role of DNA Methylation in Aortic Disease Associated With Marfan Syndrome. Cardiol Res 2025; 16:169-177. [PMID: 40370619 PMCID: PMC12074684 DOI: 10.14740/cr2033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/08/2025] [Indexed: 05/16/2025] Open
Abstract
Marfan syndrome (MFS) is a genetic disorder primarily affecting the connective tissue, with cardiovascular complications as the leading cause of mortality. While mutations in the FBN1 gene are the primary cause, the severity and progression of the disease can vary significantly among individuals. DNA methylation, a key epigenetic regulatory mechanism, has garnered attention in MFS research, particularly regarding methylation changes in the FBN1 locus and their effects on fibrillin-1 expression. Differential methylation and expression of genes related to inflammation (e.g., interleukin (IL)-10, IL-17) and oxidative stress (e.g., PON2, TP53INP1) have been linked to MFS aortic pathology. These alterations likely contribute to disease progression by influencing inflammatory responses, smooth muscle cell apoptosis, and biomechanical properties of the aorta. The transforming growth factor-beta (TGF-β) signaling pathway plays a central role in MFS pathology, with aberrant methylation of related genes potentially elevating active TGF-β levels and exacerbating aortic lesions. Notably, tissue-specific methylation patterns, especially in smooth muscle cells of the aorta, remain poorly understood. A deeper understanding of DNA methylation's role in MFS could pave the way for early interventions and epigenetic-targeted therapies.
Collapse
Affiliation(s)
- Wei Ze Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Ye Wu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Wang Z, Yang C, Dong B, Chen A, Song Q, Bai H, Jiang Y, Chang G, Chen G. Whole Transcriptome Sequencing Reveals miRNAs and ceRNA Networks in Duck Abdominal Fat Deposition. Animals (Basel) 2025; 15:506. [PMID: 40002988 PMCID: PMC11852153 DOI: 10.3390/ani15040506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Excessive deposition of abdominal fat will cause a waste of resources. In order to explore the key miRNAs and circRNA/lncRNA-miRNA-mRNA ceRNA regulatory network involved in regulating abdominal fat deposition, hematoxylin and eosin (H&E) staining was performed on abdominal fat tissues of ducks in the high abdominal fat rate group (HF) and low abdominal fat rate group (LF) at 21 and 42 days of age, and whole transcriptome sequencing was performed on abdominal tissues of ducks in the HF and LF groups at 42 days of age. The results showed that the number of adipocytes in ducks in the HF group was significantly higher than that in the LF group at 21 days of age (p < 0.001), while the number of adipocytes in ducks in the HF group at 42 days of age was significantly lower than that in the LF group (p < 0.001). In addition, transcriptome sequencing screened out a total of 14 differentially expressed miRNAs (10 miRNAs were significantly up-regulated, and 4 miRNAs were significantly down-regulated). By predicting the target genes of these differentially expressed miRNAs, a total of 305 target genes were obtained. Further analysis of miRNA target genes using GO and KEGG functional enrichment analyses revealed that these target genes were significantly enriched in the GnRH signaling pathway, the PPAR signaling pathway, insulin resistance, the mTOR signaling pathway, the AMPK signaling pathway, the FoxO signaling pathway, and other pathways related to adipose development. In addition, miRNA-205-x, miRNA-6529-x, miRNA-194-x, miRNA-215-x, miRNA-3074-x, miRNA-2954-x, novel-m0133-3p, and novel-m0156-5p were found to be important candidate miRNAs for abdominal fat deposition in ducks. These miRNAs were related to the expression of FOXO3, LIFR, Pdk4, PPARA, FBN1, MYH10, Cd44, PRELP, Esrrg, AKT3, and STC2. Based on these eight candidate miRNAs, a ceRNA regulatory network of circRNA/lncRNA-miRNA-mRNA regulating abdominal fat deposition was successfully constructed. The results of this study will provide a useful reference for accelerating the understanding of the molecular mechanism of duck abdominal fat deposition.
Collapse
|
4
|
Streubel MK, Baumgartner A, Meier-Vollrath I, Frambach Y, Brandenburger M, Kisch T. Transcriptomics of Subcutaneous Tissue of Lipedema Identified Differentially Expressed Genes Involved in Adipogenesis, Inflammation, and Pain. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e6288. [PMID: 39525887 PMCID: PMC11548906 DOI: 10.1097/gox.0000000000006288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/16/2024] [Indexed: 11/16/2024]
Abstract
Background Lipedema is a disease typically affecting women with a symmetrical, painful fat distribution disorder, which is hypothesized to be caused by impaired adipogenesis, inflammation, and extracellular matrix remodeling, leading to fibrosis and the development of edema in lipedema subcutaneous adipose tissue. The pathogenesis and molecular processes leading to lipedema have not yet been clarified. Methods A whole transcriptome analysis of subcutaneous tissue of lipedema stages I (n = 12), II (n = 9), and III (n = 8) compared with hypertrophied subcutaneous tissue (n = 4) was performed. Further data about hormonal substitution and body morphology were collected. The study is registered at ClinicalTrials.gov (NCT05861583). Results We identified several differentially expressed genes involved in mechanisms leading to the development of lipedema. Some genes, such as PRKG2, MEDAG, CSF1R, BICC1, ERBB4, and ACP5, are involved in adipogenesis, regulating the development of mature adipocytes from mesenchymal stem cells. Other genes, such as MAFB, C1Q, C2, CD68, CD209, CD163, CD84, BCAT1, and TREM2, are predicted to be involved in lipid accumulation, hypertrophy, and the inflammation process. Further genes such as SHTN1, SCN7A, and SCL12A2 are predicted to be involved in the regulation and transmission of pain. Conclusions In summary, the pathogenesis and development of lipedema might be caused by alterations in adipogenesis, inflammation, and extracellular matrix remodeling, leading to fibrosis and the formation of edema resulting in this painful disease. These processes differ from hypertrophied adipose tissue and may therefore play a main role in the formation of lipedema.
Collapse
Affiliation(s)
- Maria Karolin Streubel
- From the University of Luebeck, Luebeck, Germany
- University Hospital Schleswig-Holstein, Luebeck, Germany
| | | | | | | | - Matthias Brandenburger
- Fraunhofer Research Institution for Individualized and Cell Based Medical Engineering, IMTE, Luebeck, Germany
| | - Tobias Kisch
- From the University of Luebeck, Luebeck, Germany
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Praxisklinik Kronshagen, Kiel-Kronshagen, Germany
| |
Collapse
|
5
|
Kocaman N. Evaluating the therapeutic effect of vitamin D and nerolidol on lung injury due to experimental myocardial infarction: The potential role of asprosin and spexin. Tissue Cell 2024; 89:102444. [PMID: 38945090 DOI: 10.1016/j.tice.2024.102444] [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: 01/23/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Injury to internal organs caused by myocardial infarction (MI), although often neglected, is a very serious condition which damages internal organs especially the lungs. Changes in microcirculation can begin with acute lung injury and result in severe respiratory failure. The aim of this study was to create new approaches that will explain the pathophysiology and treatment of the disease by examining the therapeutic effects of vitamin D (VITD) and Nerolidol (NRD) on the injuries of the lungs caused by MI, and their relationship with asprosin / spexin proteins. METHODS Six groups of seven experimental animals each were constituted. Control, VITD (only 50 IU/day during the experiment), NRD (only 100 mg/kg/day during the experiment), MI (200 mg/kg isoproterenol was administered to rats as a single dose subcutaneously), MI+VITD (200 mg/kg isoproterenol +50 IU/day) and MI+NRD (200 mg/kg isoproterenol +100 mg/kg/day) were the six (6) groups constituted. Tissues were analyzed using histopathological and immunohistochemical methods, whereas serum samples were analyzed using ELISA method. RESULTS The result of the histopathological study for the MI group showed an observed increase in inflammatory cells, congestion, interalveolar septal thickening, erythrocyteloaded macrophages and fibrosis in the lung tissues. The treatment groups however recorded significant differences with regards to these parameters. In the immunohistochemical analysis, expressions of asprosin and spexin were observed in the smooth muscle structures and interalveolar areas of the vessels and bronchioles of the lung, as well as the bronchiole epithelium. There was no significant difference between the groups in terms of asprosin and spexin expression in the bronchiol epithelium. When immunohistochemical and serum ELISA results were examined, it was observed that asprosin levels increased significantly in the lung tissues of the MI group compared to the control group, decreased significantly in the treatment groups treated with Vitamin D and NRD after MI. While spexin decreased significantly in the MI group compared to the control group, it increased significantly in the MI+VİTD group, but did not change in the MI+NRD group. CONCLUSION It was observed that serious injuries occurred in the lungs due to myocardial infarction and that, VITD and NRD treatments had a curative effect on those injuries. It was also observed that Asprosin and Speksin proteins can have effect on mechanisms of both injury and therapy of the lung. Furthermore, the curative effects of VITD are dependent on the expression of asprosin and spexin; whereas the observation indicated that nerolidol could be effective through asprosin-dependent mechanisms and specisin by independent mechanisms.
Collapse
Affiliation(s)
- Nevin Kocaman
- Firat University School of Medicine, Department of Histology and Embryology Elazig, Turkey.
| |
Collapse
|
6
|
Maylem ERS, Schütz LF, Spicer LJ. The role of asprosin in regulating ovarian granulosa- and theca-cell steroidogenesis: a review with comparisons to other adipokines. Reprod Fertil Dev 2024; 36:RD24027. [PMID: 39074236 DOI: 10.1071/rd24027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024] Open
Abstract
Adipose tissues produce a variety of biologically active compounds, including cytokines, growth factors and adipokines. Adipokines are important as they function as endocrine hormones that are related to various metabolic and reproductive diseases. The goal of this review was to summarise the role of asprosin, a recently discovered adipokine, and compare its role in ovarian steroidogenesis with that of other adipokines including adiponectin, leptin, resistin, apelin, visfatin, chemerin, irisin, and gremlin 1. The summary of concentrations of these adipokines in humans, rats and other animals will help researchers identify appropriate doses to test in future studies. Review of the literature indicated that asprosin increases androstenedione production in theca cells (Tc), and when cotreated with FSH increases oestradiol production in granulosa cells (Gc). In comparison, other adipokines (1) stimulate Gc oestradiol production but inhibit Tc androgen production (adiponectin), (2) inhibit Gc oestradiol production and Tc androstenedione production (leptin and chemerin), (3) inhibit Gc steroidogenesis with no effect on Tc (resistin), (4) inhibit Gc oestradiol production but stimulate Tc androgen production (gremlin 1), and (5) increase steroid secretion by Gc, with unknown effects on Tc steroidogenesis (apelin and visfatin). Irisin has direct effects on Gc but its precise role (inhibitory or stimulatory) may be species dependent and its effects on Tc will require additional research. Thus, most adipokines have direct effects (either positive or negative) on steroid production in ovarian cells, but how they all work together to create a cumulative effect or disease will require further research.
Collapse
Affiliation(s)
- Excel Rio S Maylem
- Philippine Carabao Center, National Headquarters and Gene Pool, Science City of Munoz, Nueva Ecija, Philippines
| | - Luis Fernando Schütz
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, NV 89557, USA
| | - Leon J Spicer
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
7
|
Baban A, Parlapiano G, Cicenia M, Armando M, Franceschini A, Pacifico C, Panfili A, Zinzanella G, Romanzo A, Fusco A, Caiazza M, Perri G, Galletti L, Digilio MC, Buonuomo PS, Bartuli A, Novelli A, Raponi M, Limongelli G. Unique Features of Cardiovascular Involvement and Progression in Children with Marfan Syndrome Justify Dedicated Multidisciplinary Care. J Cardiovasc Dev Dis 2024; 11:114. [PMID: 38667733 PMCID: PMC11050181 DOI: 10.3390/jcdd11040114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Marfan syndrome (MIM: # 154700; MFS) is an autosomal dominant disease representing the most common form of heritable connective tissue disorder. The condition presents variable multiorgan expression, typically involving a triad of cardiovascular, eye, and skeletal manifestations. Other multisystemic features are often underdiagnosed. Moreover, the disease is characterized by age related penetrance. Diagnosis and management of MFS in the adult population are well-described in literature. Few studies are focused on MFS in the pediatric population, making the clinical approach (cardiac and multiorgan) to these cases challenging both in terms of diagnosis and serial follow-up. In this review, we provide an overview of MFS manifestations in children, with extensive revision of major organ involvement (cardiovascular ocular and skeletal). We attempt to shed light on minor aspects of MFS that can have a significant progressive impact on the health of affected children. MFS is an example of a syndrome where an early personalized approach to address a dynamic, genetically determined condition can make a difference in outcome. Applying an early multidisciplinary clinical approach to MFS cases can prevent acute and chronic complications, offer tailored management, and improve the quality of life of patients.
Collapse
Affiliation(s)
- Anwar Baban
- The European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart, Cardiogenetic Center, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00165 Rome, Italy; (G.P.); (A.P.)
| | - Giovanni Parlapiano
- The European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart, Cardiogenetic Center, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00165 Rome, Italy; (G.P.); (A.P.)
| | - Marianna Cicenia
- The European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart, Pediatric Cardiology and Arrhythmia/Syncope Units, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (A.F.)
| | - Michela Armando
- Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy;
| | - Alessio Franceschini
- The European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart, Pediatric Cardiology and Arrhythmia/Syncope Units, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (A.F.)
| | - Concettina Pacifico
- Audiology and Otosurgery Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Arianna Panfili
- The European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart, Cardiogenetic Center, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00165 Rome, Italy; (G.P.); (A.P.)
| | - Gaetano Zinzanella
- Ophthalmology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.Z.); (A.R.)
| | - Antonino Romanzo
- Ophthalmology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.Z.); (A.R.)
| | - Adelaide Fusco
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.F.); (M.C.); (G.L.)
| | - Martina Caiazza
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.F.); (M.C.); (G.L.)
| | - Gianluigi Perri
- Department of Pediatric Cardiology and Cardiac Surgery, Heart and Lung Transplant, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.P.); (L.G.)
| | - Lorenzo Galletti
- Department of Pediatric Cardiology and Cardiac Surgery, Heart and Lung Transplant, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.P.); (L.G.)
| | - Maria Cristina Digilio
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.D.); (P.S.B.); (A.B.)
| | - Paola Sabrina Buonuomo
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.D.); (P.S.B.); (A.B.)
| | - Andrea Bartuli
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.D.); (P.S.B.); (A.B.)
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Laboratory of Medical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Massimiliano Raponi
- Medical Direction, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Giuseppe Limongelli
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.F.); (M.C.); (G.L.)
- Centre for Paediatric Inherited and Rare Cardiovascular Disease, Institute of Cardiovascular Science, University College London, London WC1N 3JH, UK
| |
Collapse
|
8
|
Li L, Huang J, Liu Y. The extracellular matrix glycoprotein fibrillin-1 in health and disease. Front Cell Dev Biol 2024; 11:1302285. [PMID: 38269088 PMCID: PMC10806136 DOI: 10.3389/fcell.2023.1302285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
Fibrillin-1 (FBN1) is a large, cysteine-rich, calcium binding extracellular matrix glycoprotein encoded by FBN1 gene. It serves as a structural component of microfibrils and provides force-bearing mechanical support in elastic and nonelastic connective tissue. As such, mutations in the FBN1 gene can cause a wide variety of genetic diseases such as Marfan syndrome, an autosomal dominant disorder characterized by ocular, skeletal and cardiovascular abnormalities. FBN1 also interacts with numerous microfibril-associated proteins, growth factors and cell membrane receptors, thereby mediating a wide range of biological processes such as cell survival, proliferation, migration and differentiation. Dysregulation of FBN1 is involved in the pathogenesis of many human diseases, such as cancers, cardiovascular disorders and kidney diseases. Paradoxically, both depletion and overexpression of FBN1 upregulate the bioavailability and signal transduction of TGF-β via distinct mechanisms in different settings. In this review, we summarize the structure and expression of FBN1 and present our current understanding of the functional role of FBN1 in various human diseases. This knowledge will allow to develop better strategies for therapeutic intervention of FBN1 related diseases.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Junxin Huang
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
9
|
Summers KM. Genetic models of fibrillinopathies. Genetics 2024; 226:iyad189. [PMID: 37972149 PMCID: PMC11021029 DOI: 10.1093/genetics/iyad189] [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: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
The fibrillinopathies represent a group of diseases in which the 10-12 nm extracellular microfibrils are disrupted by genetic variants in one of the genes encoding fibrillin molecules, large glycoproteins of the extracellular matrix. The best-known fibrillinopathy is Marfan syndrome, an autosomal dominant condition affecting the cardiovascular, ocular, skeletal, and other systems, with a prevalence of around 1 in 3,000 across all ethnic groups. It is caused by variants of the FBN1 gene, encoding fibrillin-1, which interacts with elastin to provide strength and elasticity to connective tissues. A number of mouse models have been created in an attempt to replicate the human phenotype, although all have limitations. There are also natural bovine models and engineered models in pig and rabbit. Variants in FBN2 encoding fibrillin-2 cause congenital contractural arachnodactyly and mouse models for this condition have also been produced. In most animals, including birds, reptiles, and amphibians, there is a third fibrillin, fibrillin-3 (FBN3 gene) for which the creation of models has been difficult as the gene is degenerate and nonfunctional in mice and rats. Other eukaryotes such as the nematode C. elegans and zebrafish D. rerio have a gene with some homology to fibrillins and models have been used to discover more about the function of this family of proteins. This review looks at the phenotype, inheritance, and relevance of the various animal models for the different fibrillinopathies.
Collapse
Affiliation(s)
- Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba QLD 4102, Australia
| |
Collapse
|
10
|
Xiang Y, Wang M, Yu G, Wan L, Song Y, Li Y, Geng X, Tan L. Naringenin alleviates the excessive lipid deposition of polycystic ovary syndrome rats and insulin-resistant adipocytes by promoting PKGIα. Am J Reprod Immunol 2023; 90:e13795. [PMID: 38009056 DOI: 10.1111/aji.13795] [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: 04/04/2023] [Revised: 09/20/2023] [Accepted: 10/23/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Naringenin (NGEN) has anti-inflammatory and anti-diabetic effects. On this basis, this study aims to determine whether NGEN affects insulin resistance (IR) in polycystic ovary syndrome (PCOS). METHODS CCK-8 assay and oil red O staining were used to detect the cytotoxicity of NGEN and lipid production in cells or tissues, respectively. The differentiated mature SW872 cells were treated with palmitic acid (PA) to mimic IR cell model. Through detecting glucose consumption, the changes of inflammation and glycolipid metabolism can be observed with the assessment on expression levels of the inflammatory factors as well as lipid synthesis- (ACC, SREBP1c, PPARγ), glucose metabolism- and thermogenesis (ATGL, GLUT4, UCP1)-related genes. Insulin sensitivity was determined by changes in glucose consumption and PKGIα pathway. PKGIα was silenced to verify the protective mechanism of NGEN. PCOS rat model was constructed to confirm the results of cell experiments in vivo. RESULTS NGEN generated no effect on SW872 cell viability. SW872 cells were differentiated and mature, as evidenced by lipid droplet formation, lipid synthesis gene activation, sugar metabolism and inhibition of thermogenesis-related genes. PA induction promoted lipid synthesis in mature adipocytes, and inhibited glucose metabolism and cell insulin sensitivity. NGEN pretreatment effectively alleviated the above-mentioned abnormalities. The protective mechanism of NGEN was achieved through promoting PKGIα activation. NGEN also mitigated the abnormal glucose and lipid metabolism in PCOS rats. CONCLUSION NGEN inhibits the expression of PKGIα to alleviate IR that occurs in PCOS.
Collapse
Affiliation(s)
- Yungai Xiang
- Reproduction Centre, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Meng Wang
- Reproduction Centre, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Guo Yu
- Reproduction Centre, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Lijing Wan
- Reproduction Centre, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Yuxia Song
- Reproduction Centre, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Yan Li
- Reproduction Centre, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Xujing Geng
- Reproduction Centre, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Li Tan
- Reproduction Centre, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| |
Collapse
|
11
|
Zhang R, Guo J, Wang Y, Sun R, Dong G, Wang X, Du G. Prenatal bisphenol S exposure induces hepatic lipid deposition in male mice offspring through downregulation of adipose-derived exosomal miR-29a-3p. JOURNAL OF HAZARDOUS MATERIALS 2023; 453:131410. [PMID: 37088024 DOI: 10.1016/j.jhazmat.2023.131410] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 05/03/2023]
Abstract
The increased usage of bisphenol S (BPS) results in wide distribution in pregnant women. In this study, pregnant mice were given multiple-dose BPS during gestation. Results showed that prenatal BPS exposure (50 μg/kg/day) induced increased weight gain, dyslipidemia, higher liver triglyceride (TG), adipocyte hypertrophy, and hepatic lipid deposition in male offspring. Exosomes play important roles in regulating lipid metabolism. Here, serum exosomes and adipose miRNA sequencing of male offspring indicated a remarkable decrease in miR-29a-3p expression. To clarify whether adipocyte-derived exosomes mediate hepatic lipid deposition, exosomes were extracted from BPS-treated adipocytes and co-cultured with hepatocytes. These exosomes could be taken up by hepatocytes and promoted lipid deposition, and notably, exosomal miR-29a-3p was downregulated. Furthermore, miR-29a-3p knockdown in adipocyte-derived exosomes promoted hepatocyte lipid deposition, whereas overexpression led to the opposite effect. Also, the role of miR-29a-3p was demonstrated in hepatocytes by overexpressing or knocking it down. Subsequent studies have shown that miR-29a-3p can promote lipid deposition by directly targeting Col4a1. Taken together, prenatal BPS exposure could lead to lower miR-29a-3p yield in adipocyte-derived exosomes and decrease miR-29a-3p content transported to hepatocytes, which further negatively regulate Col4a1 and promote hepatic lipid deposition. Our findings provided clues to maternal environmental exposure-induced liver metabolic diseases.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Immunology, Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Jingyao Guo
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yupeng Wang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Rundong Sun
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Guangzhu Dong
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Baijiahu Community Health Service Center, Moling Street, Jiangning District, Nanjing 211102, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Guizhen Du
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
12
|
Ren HL, Zhao XY, Di KQ, Li LH, Hao EY, Chen H, Zhou RY, Nie CS, Wang DH. Eggshell translucency in late-phase laying hens and its effect on egg quality and physiological indicators. Front Vet Sci 2023; 10:1133752. [PMID: 37275613 PMCID: PMC10233096 DOI: 10.3389/fvets.2023.1133752] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Eggshell translucency severely affects external egg quality, and variations in the eggshell or eggshell membrane are considered the structural basis of the trait. Research has shown that 1.85% additional mixed fatty acids in the diet would greatly decrease the occurrence of eggshell translucency. Only a few studies have examined the phenotypic regularity of eggshell translucency with the increasing age of hens. Therefore, two strains, 1139 Rhode Island Red-White (RIR-White) and 836 Dwarf Layer-White (DWL-White), were used, and from each strain, 30 hens each that consecutively laid translucent or opaque eggs at 67 wks of age were selected. Subsequently, eggshell translucency, internal quality and external quality of eggs, and total cholesterol, albumin, calcium binding protein and other physiological indicators related to lipid, lipoprotein, and calcium metabolisms at the 75th, 79th, and 83rd wks of age in the late phase of the laying cycle were determined. Results: (1) In terms of flocks, for both strains, the translucency scores of the translucent groups were significantly higher than those of the opaque groups (P < 0.05); in terms of individuals, 81.1% RIR-White and 82.8% DWL-White hens consecutively laid eggs of the same or similar translucency, indicating the stability of the trait with increasing hen age; (2) In RIR-White, the eggshell strength of the translucent group at 75 weeks was significantly higher than that of the opaque group (P < 0.05); in DWL-White, the eggshell membrane thickness of the translucent group at the 75th and 83rd weeks was significantly lower than that of the opaque group (P < 0.05); (3) Compared to the opaque groups, the translucent groups had lower total cholesterol content in both RIR-White and DWL-White, lower albumin content in DWL-White at the 79th weeks (P < 0.05), and higher calcium-binding protein (CALB1) in RIR-White at the 83rd weeks (P < 0.05). In summary, this study illustrates the stability of eggshell translucency in late-phase laying hens and provides a reference of physiological indicators for exploring the formation of translucent eggs.
Collapse
Affiliation(s)
- He-Ling Ren
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Xiao-Yu Zhao
- Baoding Xingrui Agriculture and Animal Husbandry Development Co., Ltd., Baoding, China
| | - Ke-Qian Di
- School of Basic Medical Sciences, Hebei University, Baoding, China
| | - Lan-Hui Li
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Er-Ying Hao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Hui Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Rong-Yan Zhou
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Chang-Sheng Nie
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - De-He Wang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
13
|
Summers KM, Bush SJ, Davis MR, Hume DA, Keshvari S, West JA. Fibrillin-1 and asprosin, novel players in metabolic syndrome. Mol Genet Metab 2023; 138:106979. [PMID: 36630758 DOI: 10.1016/j.ymgme.2022.106979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Fibrillin-1 is a major component of the extracellular microfibrils, where it interacts with other extracellular matrix proteins to provide elasticity to connective tissues, and regulates the bioavailability of TGFβ family members. A peptide consisting of the C-terminal 140 amino acids of fibrillin-1 has recently been identified as a glucogenic hormone, secreted from adipose tissue during fasting and targeting the liver to release glucose. This fragment, called asprosin, also signals in the hypothalamus to stimulate appetite. Asprosin levels are correlated with many of the pathologies indicative of metabolic syndrome, including insulin resistance and obesity. Previous studies and reviews have addressed the therapeutic potential of asprosin as a target in obesity, diabetes and related conditions without considering mechanisms underlying the relationship between generation of asprosin and expression of the much larger fibrillin-1 protein. Profibrillin-1 undergoes obligatory cleavage at the cell surface as part of its assembly into microfibrils, producing the asprosin peptide as well as mature fibrillin-1. Patterns of FBN1 mRNA expression are inconsistent with the necessity for regulated release of asprosin. The asprosin peptide may be protected from degradation in adipose tissue. We present evidence for an alternative possibility, that asprosin mRNA is generated independently from an internal promoter within the 3' end of the FBN1 gene, which would allow for regulation independent of fibrillin-synthesis and is more economical of cellular resources. The discovery of asprosin opened exciting possibilities for treatment of metabolic syndrome related conditions, but there is much to be understood before such therapies could be introduced into the clinic.
Collapse
Affiliation(s)
- Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, Queensland 4102, Australia.
| | - Stephen J Bush
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, United Kingdom.
| | - Margaret R Davis
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, Queensland 4102, Australia.
| | - Sahar Keshvari
- Mater Research Institute-University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, Queensland 4102, Australia.
| | - Jennifer A West
- Faculty of Medicine, The University of Queensland, Mayne Medical Building, 288 Herston Road, Herston, Queensland 4006, Australia.
| |
Collapse
|
14
|
Tiedemann K, Muthu ML, Reinhardt DP, Komarova SV. Male Marfan mice are predisposed to high fat diet induced obesity, diabetes, and fatty liver. Am J Physiol Cell Physiol 2022; 323:C354-C366. [PMID: 35759435 DOI: 10.1152/ajpcell.00062.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gene mutations in the extracellular matrix protein fibrillin-1 cause connective tissue disorders including Marfan syndrome (MFS) with clinical symptoms in the cardiovascular, skeletal, and ocular systems. MFS patients also exhibit alterations in adipose tissues, which in some individuals leads to lipodystrophy, whereas in others to obesity. We have recently demonstrated that fibrillin-1 regulates adipose tissue homeostasis. Here, we examined how fibrillin-1 abnormality affects metabolic adaptation to different diets. We used two MFS mouse models: Hypomorph Fbn1mgR/mgR mice and Fbn1C1041G/+ mice with a fibrillin-1 missense mutation. When Fbn1mgR/mgR mice were fed with high fat diet (HFD) for 12 weeks, male mice were heavier than littermate controls (LC), whereas female mice gained less weight compared to LC. Female Fbn1C1041G/+ mice on a HFD for 24 weeks were similarly protected from weight gain. Male Fbn1C1041G/+ mice on HFD demonstrated higher insulin levels, insulin intolerance, circulating levels of cholesterol and high-density lipoproteins. Moreover, male HFD-fed Fbn1C1041G/+ mice showed a higher liver weight and a fatty liver phenotype, which was reduced to LC levels after orchiectomy. Phosphorylation of protein kinase-like endoplasmic reticulum kinase (PERK) as well as the expression of sterol regulatory element-binding protein 1 (Srebp1) in livers of HFD-fed male Fbn1C1041G/+ mice were elevated. In conclusion, the data demonstrate that male mice of both MFS models are susceptible to HFD-induced obesity and diabetes. Moreover, male Fbn1C1041G/+ mice develop a fatty liver phenotype, likely mediated by a baseline increased endoplasmic reticulum stress. In contrast, female MFS mice were protected from the consequence of HFD.
Collapse
Affiliation(s)
- Kerstin Tiedemann
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, Canada.,Shriners Hospital for Children - Canada, Montréal, Canada
| | - Muthu L Muthu
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Dieter P Reinhardt
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, Canada.,Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Svetlana V Komarova
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, Canada.,Shriners Hospital for Children - Canada, Montréal, Canada
| |
Collapse
|
15
|
Two Novel Biomarker Candidates for Differentiating Basal Cell Carcinoma from Trichoblastoma; Asprosin and Meteorine Like Peptide. Tissue Cell 2022; 76:101752. [DOI: 10.1016/j.tice.2022.101752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 01/08/2023]
|
16
|
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]
|
17
|
Summers KM, Bush SJ, Wu C, Hume DA. Generation and network analysis of an RNA-seq transcriptional atlas for the rat. NAR Genom Bioinform 2022; 4:lqac017. [PMID: 35265836 PMCID: PMC8900154 DOI: 10.1093/nargab/lqac017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/13/2022] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
Abstract
The laboratory rat is an important model for biomedical research. To generate a comprehensive rat transcriptomic atlas, we curated and downloaded 7700 rat RNA-seq datasets from public repositories, downsampled them to a common depth and quantified expression. Data from 585 rat tissues and cells, averaged from each BioProject, can be visualized and queried at http://biogps.org/ratatlas. Gene co-expression network (GCN) analysis revealed clusters of transcripts that were tissue or cell type restricted and contained transcription factors implicated in lineage determination. Other clusters were enriched for transcripts associated with biological processes. Many of these clusters overlap with previous data from analysis of other species, while some (e.g. expressed specifically in immune cells, retina/pineal gland, pituitary and germ cells) are unique to these data. GCN analysis on large subsets of the data related specifically to liver, nervous system, kidney, musculoskeletal system and cardiovascular system enabled deconvolution of cell type-specific signatures. The approach is extensible and the dataset can be used as a point of reference from which to analyse the transcriptomes of cell types and tissues that have not yet been sampled. Sets of strictly co-expressed transcripts provide a resource for critical interpretation of single-cell RNA-seq data.
Collapse
Affiliation(s)
- Kim M Summers
- Mater Research Institute—University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, QLD 4102, Australia
| | - Stephen J Bush
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Chunlei Wu
- Department of Integrative and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David A Hume
- Mater Research Institute—University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
18
|
Matrisome alterations in obesity – Adipose tissue transcriptome study on monozygotic weight-discordant twins. Matrix Biol 2022; 108:1-19. [DOI: 10.1016/j.matbio.2022.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 12/11/2022]
|
19
|
Shabir K, Brown JE, Afzal I, Gharanei S, Weickert MO, Barber TM, Kyrou I, Randeva HS. Asprosin, a novel pleiotropic adipokine implicated in fasting and obesity-related cardio-metabolic disease: Comprehensive review of preclinical and clinical evidence. Cytokine Growth Factor Rev 2021; 60:120-132. [PMID: 34108103 DOI: 10.1016/j.cytogfr.2021.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/09/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022]
Abstract
White adipose tissue is a dynamic endocrine organ that releases an array of adipokines, which play a key role in regulating metabolic homeostasis and multiple other physiological processes. An altered adipokine secretion profile from adipose tissue depots frequently characterizes obesity and related cardio-metabolic diseases. Asprosin is a recently discovered adipokine that is released in response to fasting. Following secretion, asprosin acts - via an olfactory G-protein coupled receptor and potentially via other unknown receptor(s) - on hepatocytes and agouti-related peptide-expressing neurons in the central nervous system to stimulate glucose secretion and promote appetite, respectively. A growing body of both in vitro and in vivo studies have shown asprosin to exert a number of effects on different metabolic tissues. Indeed, asprosin can attenuate insulin signalling and promote insulin resistance in skeletal muscle by increasing inflammation and endoplasmic reticulum stress. Interestingly, asprosin may also play a protective role in cardiomyocytes that are exposed to hypoxic conditions. Moreover, clinical studies have reported elevated circulating asprosin levels in obesity, type 2 diabetes and other obesity-related cardio-metabolic diseases, with significant associations to clinically relevant parameters. Understanding the spectrum of the effects of this novel adipokine is essential in order to determine its physiologic role and its significance as a potential therapeutic target and/or a biomarker of cardio-metabolic disease. The present review offers a comprehensive overview of the published literature on asprosin, including both clinical and preclinical studies, focusing on its role in metabolism and cardio-metabolic disease.
Collapse
Affiliation(s)
- Kiran Shabir
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom
| | - James E Brown
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom; School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom
| | - Islam Afzal
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom
| | - Seley Gharanei
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Martin O Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom; Centre of Applied Biological & Exercise Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry, CV1 5FB, United Kingdom
| | - Thomas M Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom; Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, CV1 5FB, United Kingdom.
| | - Harpal S Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| |
Collapse
|
20
|
A phenome-wide association study of 26 mendelian genes reveals phenotypic expressivity of common and rare variants within the general population. PLoS Genet 2020; 16:e1008802. [PMID: 33226994 PMCID: PMC7735621 DOI: 10.1371/journal.pgen.1008802] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/14/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
The clinical evaluation of a genetic syndrome relies upon recognition of a characteristic pattern of signs or symptoms to guide targeted genetic testing for confirmation of the diagnosis. However, individuals displaying a single phenotype of a complex syndrome may not meet criteria for clinical diagnosis or genetic testing. Here, we present a phenome-wide association study (PheWAS) approach to systematically explore the phenotypic expressivity of common and rare alleles in genes associated with four well-described syndromic diseases (Alagille (AS), Marfan (MS), DiGeorge (DS), and Noonan (NS) syndromes) in the general population. Using human phenotype ontology (HPO) terms, we systematically mapped 60 phenotypes related to AS, MS, DS and NS in 337,198 unrelated white British from the UK Biobank (UKBB) based on their hospital admission records, self-administrated questionnaires, and physiological measurements. We performed logistic regression adjusting for age, sex, and the first 5 genetic principal components, for each phenotype and each variant in the target genes (JAG1, NOTCH2 FBN1, PTPN1 and RAS-opathy genes, and genes in the 22q11.2 locus) and performed a gene burden test. Overall, we observed multiple phenotype-genotype correlations, such as the association between variation in JAG1, FBN1, PTPN11 and SOS2 with diastolic and systolic blood pressure; and pleiotropy among multiple variants in syndromic genes. For example, rs11066309 in PTPN11 was significantly associated with a lower body mass index, an increased risk of hypothyroidism and a smaller size for gestational age, all in concordance with NS-related phenotypes. Similarly, rs589668 in FBN1 was associated with an increase in body height and blood pressure, and a reduced body fat percentage as observed in Marfan syndrome. Our findings suggest that the spectrum of associations of common and rare variants in genes involved in syndromic diseases can be extended to individual phenotypes within the general population. Standard medical evaluation of genetic syndromes relies upon recognizing a characteristic pattern of signs or symptoms to guide targeted genetic testing for confirmation of the diagnosis. This may lead to missing diagnoses in patients with silent or a low expressed form of the syndrome. Here we take advantage of a rich electronic health record, various phenotypic measurements, and genetic information in 337,198 unrelated white British from the UKBB, to study the relation between single syndromic disease phenotypes and genes related to syndromic disease. We show multiple phenotype-genotype associations in concordance with phenotypes variations found in syndromic diseases. For example, we show that a commonly found variant in FBN1 was associated with high standing/sitting height ratio and reduced body fat percentage as observed in individuals with Marfan syndrome. Our findings suggest that common and rare alleles in syndromic disease genes are causative of individual component phenotypes present in a general population; further research is needed to characterize the pleiotropic effect of alleles in syndromic genes in persons without the syndromic disease.
Collapse
|
21
|
Mohiuddin OA, Motherwell JM, Rogers E, Bratton MR, Zhang Q, Wang G, Bunnell B, Hayes DJ, Gimble JM. Characterization and Proteomic Analysis of Decellularized Adipose Tissue Hydrogels Derived from Lean and Overweight/Obese Human Donors. ACTA ACUST UNITED AC 2020; 4:e2000124. [PMID: 32914579 DOI: 10.1002/adbi.202000124] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/28/2020] [Indexed: 12/19/2022]
Abstract
While decellularized adipose tissue (DAT) has potential as an "off-the-shelf" biomaterial product for regenerative medicine, it remains to be determined if donor-source body mass index (BMI) impacts the functionality of DAT. This study set out to comparatively characterize lean versus overweight/obese-donor derived DAT hydrogel based on proteome and to analyze their respective effects on adipose stromal/stem cell (ASC) viability, and differentiation in vitro. Decellularized adipose tissue from lean (lDAT) and overweight/obese (oDAT) donors is produced and characterized. Variability in the fibril microstructures is found, with dense fibrotic fiber clusters and large pore area uniquely present in the oDAT samples. Proteomic analysis reveals that lDAT contains a greater proportion of enriched extracellular proteins and a smaller proportion of enriched intracellular proteins relative to oDAT. Biocompatibility studies show that ASCs cultured in lDAT and oDAT hydrogels remain viable. The adipogenic and osteogenic differentiation capability of ASCs seeded in lDAT and oDAT hydrogels is confirmed by an upregulation in marker gene expression and phenotypic analysis. In conclusion, this study establishes that DAT hydrogels derived from lean and overweight/obese adipose donors present similar physicochemical profiles with some distinctive features while comparably supporting the viability and adipogenic differentiation of ASCs in vitro.
Collapse
Affiliation(s)
- Omair A Mohiuddin
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Jessica M Motherwell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Emma Rogers
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70112, USA
| | | | - Qiang Zhang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | - Guangdi Wang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | - Bruce Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, Pennsylvania State University, State College, PA, 16802, USA
| | - Jeffrey M Gimble
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- LaCell LLC and Obatala Sciences Inc., New Orleans, LA, 70148, USA
| |
Collapse
|
22
|
Wang T, Hill RC, Dzieciatkowska M, Zhu L, Infante AM, Hu G, Hansen KC, Pei M. Site-Dependent Lineage Preference of Adipose Stem Cells. Front Cell Dev Biol 2020; 8:237. [PMID: 32351957 PMCID: PMC7174673 DOI: 10.3389/fcell.2020.00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
Adult stem cells have unique properties in both proliferation and differentiation preference. In this study, we hypothesized that adipose stem cells have a depot-dependent lineage preference. Four rabbits were used to provide donor-matched adipose stem cells from either subcutaneous adipose tissue (ScAT) or infrapatellar fat pad (IPFP). Proliferation and multi-lineage differentiation were evaluated in adipose stem cells from donor-matched ScAT and IPFP. RNA sequencing (RNA-seq) and proteomics were conducted to uncover potential molecular discrepancy in adipose stem cells and their corresponding matrix microenvironments. We found that stem cells from ScAT exhibited significantly higher proliferation and adipogenic capacity compared to those from donor-matched IPFP while stem cells from IPFP displayed significantly higher chondrogenic potential compared to those from donor-matched ScAT. Our findings are strongly endorsed by supportive data from transcriptome and proteomics analyses, indicating a site-dependent lineage preference of adipose stem cells.
Collapse
Affiliation(s)
- Tingliang Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopedics, West Virginia University, Morgantown, WV, United States
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ryan C. Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aniello M. Infante
- Bioinformatics Core Facility, West Virginia University, Morgantown, WV, United States
| | - Gangqing Hu
- Bioinformatics Core Facility, West Virginia University, Morgantown, WV, United States
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopedics, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
23
|
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
|
24
|
Csala A, Zwinderman AH, Hof MH. Multiset sparse partial least squares path modeling for high dimensional omics data analysis. BMC Bioinformatics 2020; 21:9. [PMID: 31918677 PMCID: PMC6953292 DOI: 10.1186/s12859-019-3286-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 11/20/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Recent technological developments have enabled the measurement of a plethora of biomolecular data from various omics domains, and research is ongoing on statistical methods to leverage these omics data to better model and understand biological pathways and genetic architectures of complex phenotypes. Current reviews report that the simultaneous analysis of multiple (i.e. three or more) high dimensional omics data sources is still challenging and suitable statistical methods are unavailable. Often mentioned challenges are the lack of accounting for the hierarchical structure between omics domains and the difficulty of interpretation of genomewide results. This study is motivated to address these challenges. We propose multiset sparse Partial Least Squares path modeling (msPLS), a generalized penalized form of Partial Least Squares path modeling, for the simultaneous modeling of biological pathways across multiple omics domains. msPLS simultaneously models the effect of multiple molecular markers, from multiple omics domains, on the variation of multiple phenotypic variables, while accounting for the relationships between data sources, and provides sparse results. The sparsity in the model helps to provide interpretable results from analyses of hundreds of thousands of biomolecular variables. RESULTS With simulation studies, we quantified the ability of msPLS to discover associated variables among high dimensional data sources. Furthermore, we analysed high dimensional omics datasets to explore biological pathways associated with Marfan syndrome and with Chronic Lymphocytic Leukaemia. Additionally, we compared the results of msPLS to the results of Multi-Omics Factor Analysis (MOFA), which is an alternative method to analyse this type of data. CONCLUSIONS msPLS is an multiset multivariate method for the integrative analysis of multiple high dimensional omics data sources. It accounts for the relationship between multiple high dimensional data sources while it provides interpretable results through its sparse solutions. The biomarkers found by msPLS in the omics datasets can be interpreted in terms of biological pathways associated with the pathophysiology of Marfan syndrome and of Chronic Lymphocytic Leukaemia. Additionally, msPLS outperforms MOFA in terms of variation explained in the chronic lymphocytic leukaemia dataset while it identifies the two most important clinical markers for Chronic Lymphocytic Leukaemia AVAILABILITY: http://uva.csala.me/mspls.https://github.com/acsala/2018_msPLS.
Collapse
Affiliation(s)
- Attila Csala
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, 1105 AZ The Netherlands
| | - Aeilko H. Zwinderman
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, 1105 AZ The Netherlands
| | - Michel H. Hof
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, 1105 AZ The Netherlands
| |
Collapse
|
25
|
Lin M, Liu Z, Liu G, Zhao S, Li C, Chen W, Coban Akdemir Z, Lin J, Song X, Wang S, Xu Q, Zhao Y, Wang L, Zhang Y, Yan Z, Liu S, Liu J, Chen Y, Zuo Y, Yang X, Sun T, Yang X, Niu Y, Li X, You W, Qiu B, Ding C, Liu P, Zhang S, Carvalho CMB, Posey JE, Qiu G, Deciphering Disorders Involving Scoliosis and COmorbidities (DISCO) study, Lupski JR, Wu Z, Zhang J, Wu N. Genetic and molecular mechanism for distinct clinical phenotypes conveyed by allelic truncating mutations implicated in FBN1. Mol Genet Genomic Med 2020; 8:e1023. [PMID: 31774634 PMCID: PMC6978264 DOI: 10.1002/mgg3.1023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/21/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The molecular and genetic mechanisms by which different single nucleotide variant alleles in specific genes, or at the same genetic locus, cause distinct disease phenotypes often remain unclear. Allelic truncating mutations of FBN1 could cause either classical Marfan syndrome (MFS) or a more complicated phenotype associated with Marfanoid-progeroid-lipodystrophy syndrome (MPLS). METHODS We investigated a small cohort, encompassing two classical MFS and one MPLS subjects from China, whose clinical presentation included scoliosis potentially requiring surgical intervention. Targeted next generation sequencing was performed on all the participants. We analyzed the molecular diagnosis, clinical features, and the potential molecular mechanism involved in the MPLS subject in our cohort. RESULTS We report a novel de novo FBN1 mutation for the first Chinese subject with MPLS, a more complicated fibrillinopathy, and two subjects with more classical MFS. We further predict that the MPLS truncating mutation, and others previously reported, is prone to escape the nonsense-mediated decay (NMD), while MFS mutations are predicted to be subjected to NMD. Also, the MPLS mutation occurs within the glucogenic hormone asprosin domain of FBN1. In vitro experiments showed that the single MPLS mutation p.Glu2759Cysfs*9 appears to perturb proper FBN1 protein aggregation as compared with the classical MFS mutation p.Tyr2596Thrfs*86. Both mutations appear to upregulate SMAD2 phosphorylation in vitro. CONCLUSION We provide direct evidence that a dominant-negative interaction of FBN1 potentially explains the complex MPLS phenotypes through genetic and functional analysis. Our study expands the mutation spectrum of FBN1 and highlights the potential molecular mechanism for MPLS.
Collapse
|
26
|
Wiecek M, Szymura J, Sproull J, Szygula Z. Decreased Blood Asprosin in Hyperglycemic Menopausal Women as a Result of Whole-Body Cryotherapy Regardless of Metabolic Syndrome. J Clin Med 2019; 8:E1428. [PMID: 31510055 PMCID: PMC6780623 DOI: 10.3390/jcm8091428] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/26/2022] Open
Abstract
Endocrine dysfunction often occurs in metabolic syndrome (MetS), resulting in hyperglycemia and atherogenic blood lipid profile disorders. Asprosin is a newly discovered glucose-regulating hormone. The study aim was to determine whether the application of whole-body cryotherapy (WBC) affects asprosin and selected adipocytokines as well as insulin resistance in menopausal women with metabolic disorders. A total of 37 menopausal women were exposed to 20 WBC (-130 °C, 3 min). Blood glucose, asprosin, irisin, leptin, adiponectin, and insulin were measured before and after 20 WBC treatments, after which a homeostasis model assessment of insulin resistance (HOMA-IR) and atherogenic index of plasma (AIP) were calculated. The results were analyzed in the MetS group compared to the controls (CON) without MetS, and in the hyperglycemic (HG) group compared to the normoglycemic group (NG). After 20 WBC, a significant reduction (p < 0.05) in asprosin concentration was found in the MetS, HG, and CON groups, and a significant decrease (p < 0.05) in glucose concentration was noted in the HG group. Changes in asprosin concentration positively correlated with changes in glucose concentration. Asprosin concentration before WBC correlated positively with metabolic disorder risk factor levels, and the change in asprosin concentration after 20 WBC correlated negatively with metabolic disorder risk factor levels: fasting glucose, AIP, and the leptin/adiponectin index. Research indicates the possibility of using WBC in supporting metabolic disorders, type 2 diabetes (T2DM), and insulin resistance.
Collapse
Affiliation(s)
- Magdalena Wiecek
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, University of Physical Education in Krakow, 31-571 Krakow, Poland.
| | - Jadwiga Szymura
- Department of Clinical Rehabilitation, Faculty of Motor Rehabilitation, University of Physical Education in Krakow, 31-571 Krakow, Poland.
| | - Justyna Sproull
- Faculty of Physical Education and Sport, University of Physical Education in Krakow, 31-571 Krakow, Poland.
| | - Zbigniew Szygula
- Institute of Health Sciences, State Higher Vocational School in Tarnow, 33-100 Tarnow, Poland.
| |
Collapse
|
27
|
Yu B, Peng XH, Wang LY, Wang AB, Su YY, Chen JH, Zhang XW, Zhao DZ, Wang H, Pang DX, Ouyang HS, Tang XC, Zhang MJ. Abnormality of intestinal cholesterol absorption in ApcMin/+ mice with colon cancer cachexia. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:759-767. [PMID: 31933883 PMCID: PMC6945172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 01/24/2019] [Indexed: 06/10/2023]
Abstract
Colorectal cancer syndrome has been one of the greatest concerns in the world, particularly in developed countries. Several epidemiological studies have shown that dyslipidemia may be associated with the progression of intestinal cachexia, but there is little research on the function of the small intestine, which is involved in blood lipid metabolism, in dyslipidemia. In the present study, we aimed to explore the function of intestinal cholesterol absorption in the ApcMin/+ mouse model using an intestinal lipid absorption test. We found that both triglyceride (TG) and total cholesterol (TC) uptake were inhibited in the intestine of ApcMin/+ mice with age and the intestinal peroxisome proliferator-activated receptor α (PPARα) downregulated the processes of β-oxidation, oxidative stress response, and cholesterol absorption in APC-deficient mice. In addition, reduced expression levels of farnesoid X receptor (FXR) and apical sodium-dependent bile acid transporter (ASBT) indicated that bile acid metabolism might be associated with intestinal cholesterol absorption in ApcMin/+ mice. Thus, our data suggested that the intestine plays an essential role in cholesterol uptake and that bile acid metabolism seems to cause a decrease in intestinal cholesterol uptake in ApcMin/+ mice.
Collapse
Affiliation(s)
- Biao Yu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Xiao-Huan Peng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Ling-Yu Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - An-Bei Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Yan-Yan Su
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Jia-Huan Chen
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Xin-Wei Zhang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Da-Zhong Zhao
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - He Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Da-Xin Pang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Hong-Sheng Ouyang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Xiao-Chun Tang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Ming-Jun Zhang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| |
Collapse
|
28
|
Coles CA, Maksimovic J, Wadeson J, Fahri FT, Webster T, Leyton C, McDonagh MB, White JD. Knockdown of a disintegrin A metalloprotease 12 (ADAM12) during adipogenesis reduces cell numbers, delays differentiation, and increases lipid accumulation in 3T3-L1 cells. Mol Biol Cell 2018; 29:1839-1855. [PMID: 29846135 PMCID: PMC6085825 DOI: 10.1091/mbc.e17-07-0471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mouse models have shown that a disintegrin A metalloprotease 12 (ADAM12) is implicated during adipogenesis; the molecular pathways are not well understood. Stealth RNA interference was used to knock down ADAM12 in 3T3-L1 cells. Using gene profiling and metabolic enzymatic markers, we have identified signaling pathways ADAM12 impacts upon during proliferation, differentiation, and maturation of adipocytes. ADAM12 reduced cell numbers in proliferating preadipocytes, delayed differentiation of preadipocytes to adipocytes, and increased lipid accumulation in mature adipocytes. The pathway most affected by ADAM12 knockdown was regulation of insulin-like growth factor (IGF) activity by insulin-like growth factor binding proteins (IGFBPs); ADAM12 is known to cleave IGFBP3 and IGFBP5. The IGF/mTOR signaling pathway was down-regulated, supporting a role for ADAM12 in the IGFBP/IGF/mTOR-growth pathway. PPARγ signaling was also down-regulated by ADAM12 knockdown. Gene ontology (GO) analysis revealed that the extracellular matrix was the cellular compartment most impacted. Filtering for matrisome genes, connective tissue growth factor (Ctgf) was up-regulated. CTGF and IGBP3 can interact with PPARγ to hinder its regulation. Increased expression of these molecules could have influenced PPARγ signaling reducing differentiation and an imbalance of lipids. We believe ADAM12 regulates cell proliferation of preadipocytes through IGFBP/IGF/mTOR signaling and delays differentiation through altered PPAR signaling to cause an imbalance of lipids within mature adipocytes.
Collapse
Affiliation(s)
- Chantal A Coles
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3052, Australia.,Faculty of Veterinary and Agricultural Science, University of Melbourne, Melbourne 3052, Australia
| | - Jovana Maksimovic
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne 3052, Australia
| | - Jenny Wadeson
- Discovery Technologies, Department of Environment and Primary -Industries, Victoria Centre for AgriBioscience, La Trobe University, Melbourne 3083, Australia
| | - Fahri T Fahri
- Department of Primary Industries, New South Wales Food Authority, Sydney, New South Wales 2001, Australia
| | - Tracie Webster
- Discovery Technologies, Department of Environment and Primary -Industries, Victoria Centre for AgriBioscience, La Trobe University, Melbourne 3083, Australia
| | - Carolina Leyton
- Discovery Technologies, Department of Environment and Primary -Industries, Victoria Centre for AgriBioscience, La Trobe University, Melbourne 3083, Australia
| | - Matthew B McDonagh
- Discovery Technologies, Department of Environment and Primary -Industries, Victoria Centre for AgriBioscience, La Trobe University, Melbourne 3083, Australia.,Cooperative Animal Research Centre for Sheep Industry Innovation, University of New England, Armidale, New South Wales 2350, Australia
| | - Jason D White
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3052, Australia.,Faculty of Veterinary and Agricultural Science, University of Melbourne, Melbourne 3052, Australia
| |
Collapse
|
29
|
Chen M, Yao B, Yang Q, Deng J, Song Y, Sui T, Zhou L, Yao H, Xu Y, Ouyang H, Pang D, Li Z, Lai L. Truncated C-terminus of fibrillin-1 induces Marfanoid-progeroid-lipodystrophy (MPL) syndrome in rabbit. Dis Model Mech 2018; 11:dmm.031542. [PMID: 29666143 PMCID: PMC5963856 DOI: 10.1242/dmm.031542] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/05/2018] [Indexed: 12/12/2022] Open
Abstract
Various clinical differences have been observed between patients with the FBN1 gene mutation and those with the classical Marfan phenotype. Although FBN1 knockout (KO) or dominant-negative mutant mice are widely used as an animal model for Marfan syndrome (MFS), these mice cannot recapitulate the genotype/phenotype relationship of Marfanoid-progeroid-lipodystrophy (MPL) syndrome, which is caused by a mutation in the C-terminus of fibrillin-1, the penultimate exon of the FBN1 gene. Here, we describe the generation of a rabbit MPL model with C-terminal truncation of fibrillin-1 using a CRISPR/Cas9 system. FBN1 heterozygous (FBN1 Het) rabbits faithfully recapitulated the phenotypes of MFS, including muscle wasting and impaired connective tissue, ocular syndrome and aortic dilation. Moreover, skin symptoms, lipodystrophy, growth retardation and dysglycemia were also seen in these FBN1 Het rabbits, and have not been reported in other animal models. In conclusion, this novel rabbit model mimics the histopathological changes and functional defects of MPL syndrome, and could become a valuable model for studies of pathogenesis and drug screening for MPL syndrome. Summary: A novel genetically engineered rabbit model of MPL syndrome, generated by CRISPR/Cas9-mediated mutation of FBN1, mimics the histopathological changes and functional defects of MPL syndrome seen in the clinic.
Collapse
Affiliation(s)
- Mao Chen
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Bing Yao
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Qiangbing Yang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Jichao Deng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Yuning Song
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Tingting Sui
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Lina Zhou
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - HaoBing Yao
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Yuanyuan Xu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Hongsheng Ouyang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Daxin Pang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Zhanjun Li
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China
| | - Liangxue Lai
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China .,Key Laboratory of Regenerative Biology, Chinese Academy of Sciences, and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| |
Collapse
|
30
|
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
|
31
|
Davis MR, Arner E, Duffy CRE, De Sousa PA, Dahlman I, Arner P, Summers KM. Datasets of genes coexpressed with FBN1 in mouse adipose tissue and during human adipogenesis. Data Brief 2016; 8:851-7. [PMID: 27508231 PMCID: PMC4959917 DOI: 10.1016/j.dib.2016.06.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 12/22/2022] Open
Abstract
This article contains data related to the research article entitled “Expression of FBN1 during adipogenesis: relevance to the lipodystrophy phenotype in Marfan syndrome and related conditions” [1]. The article concerns the expression of FBN1, the gene encoding the extracellular matrix protein fibrillin-1, during adipogenesis in vitro and in relation to adipose tissue in vivo. The encoded protein has recently been shown to produce a short glucogenic peptide hormone, (Romere et al., 2016) [2], and this gene is therefore a key gene for regulating blood glucose levels. FBN1 and coexpressed genes were examined in mouse strains and in human cells undergoing adipogenesis. The data show the genes that were coexpressed with FBN1, including genes coding for other connective tissue proteins and the proteases that modify them and for the transcription factors that control their expression. Data analysed were derived from datasets available in the public domain and the analysis highlights the utility of such datasets for ongoing analysis and hence reduction in the use of experimental animals.
Collapse
Affiliation(s)
- Margaret R Davis
- The Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Erik Arner
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies) (CLST (DGT)), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Cairnan R E Duffy
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellors Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Paul A De Sousa
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellors Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Ingrid Dahlman
- Department of Medicine, Huddinge (Med H), Karolinska Universitetssjukhuset Huddinge, 141 86 Stockholm, Sweden
| | - Peter Arner
- Department of Medicine, Huddinge (Med H), Karolinska Universitetssjukhuset Huddinge, 141 86 Stockholm, Sweden
| | - Kim M Summers
- The Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| |
Collapse
|