1
|
Zhang E, Yan Q, Sun Y, Li J, Chen L, Zou J, Zeng S, Jiang J, Li J. Integrative Analysis of Lactylome and Proteome of Hypertrophic Scar To Identify Pathways or Proteins Associated with Disease Development. J Proteome Res 2024; 23:3367-3382. [PMID: 39012622 DOI: 10.1021/acs.jproteome.3c00901] [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] [Indexed: 07/17/2024]
Abstract
Lactylation (Kla), a recently discovered post-translational modification derived from lactate, plays crucial roles in various cellular processes. However, the specific influence of lactylation on the biological processes underlying hypertrophic scar formation remains unclear. In this study, we present a comprehensive profiling of the lactylome and proteome in both hypertrophic scars and adjacent normal skin tissues. A total of 1023 Kla sites originating from 338 nonhistone proteins were identified based on lactylome analysis. Proteome analysis in hypertrophic scar and adjacent skin samples revealed the identification of 2008 proteins. It is worth noting that Kla exhibits a preference for genes associated with ribosome function as well as glycolysis/gluconeogenesis in both normal skin and hypertrophic scar tissues. Furthermore, the functional enrichment analysis demonstrated that differentially lactyled proteins are primarily involved in proteoglycans, HIF-1, and AMPK signaling pathways. The combined analysis of the lactylome and proteome data highlighted a significant upregulation of 14 lactylation sites in hypertrophic scar tissues. Overall, our investigation unveiled the significant involvement of protein lactylation in the regulation of ribosome function as well as glycolysis/gluconeogenesis, potentially contributing to the formation of hypertrophic scars.
Collapse
Affiliation(s)
- Enyuan Zhang
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Qiyue Yan
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Yue Sun
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jingyun Li
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Ling Chen
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jijun Zou
- Department of Burns and Plastic Surgery, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| | - Siqi Zeng
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jingbin Jiang
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jun Li
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| |
Collapse
|
2
|
Lyons PJ. Inactive metallopeptidase homologs: the secret lives of pseudopeptidases. Front Mol Biosci 2024; 11:1436917. [PMID: 39050735 PMCID: PMC11266112 DOI: 10.3389/fmolb.2024.1436917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Inactive enzyme homologs, or pseudoenzymes, are proteins, found within most enzyme families, that are incapable of performing catalysis. Rather than catalysis, they are involved in protein-protein interactions, sometimes regulating the activity of their active enzyme cousins, or scaffolding protein complexes. Pseudoenzymes found within metallopeptidase families likewise perform these functions. Pseudoenzymes within the M14 carboxypeptidase family interact with collagens within the extracellular space, while pseudopeptidase members of the M12 "a disintegrin and metalloprotease" (ADAM) family either discard their pseudopeptidase domains as unnecessary for their roles in sperm maturation or utilize surface loops to enable assembly of key complexes at neuronal synapses. Other metallopeptidase families contain pseudopeptidases involved in protein synthesis at the ribosome and protein import into organelles, sometimes using their pseudo-active sites for these interactions. Although the functions of these pseudopeptidases have been challenging to study, ongoing work is teasing out the secret lives of these proteins.
Collapse
Affiliation(s)
- Peter J. Lyons
- Department of Biology, Andrews University, Berrien Springs, MI, United States
| |
Collapse
|
3
|
Shestakova A, Fatkulin A, Surkova D, Osmolovskiy A, Popova E. First Insight into the Degradome of Aspergillus ochraceus: Novel Secreted Peptidases and Their Inhibitors. Int J Mol Sci 2024; 25:7121. [PMID: 39000228 PMCID: PMC11241649 DOI: 10.3390/ijms25137121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Aspergillus fungi constitute a pivotal element within ecosystems, serving as both contributors of biologically active compounds and harboring the potential to cause various diseases across living organisms. The organism's proteolytic enzyme complex, termed the degradome, acts as an intermediary in its dynamic interaction with the surrounding environment. Using techniques such as genome and transcriptome sequencing, alongside protein prediction methodologies, we identified putative extracellular peptidases within Aspergillus ochraceus VKM-F4104D. Following manual annotation procedures, a total of 11 aspartic, 2 cysteine, 2 glutamic, 21 serine, 1 threonine, and 21 metallopeptidases were attributed to the extracellular degradome of A. ochraceus VKM-F4104D. Among them are enzymes with promising applications in biotechnology, potential targets and agents for antifungal therapy, and microbial antagonism factors. Thus, additional functionalities of the extracellular degradome, extending beyond mere protein substrate digestion for nutritional purposes, were demonstrated.
Collapse
Affiliation(s)
- Anna Shestakova
- Department of Microbiology, Lomonosov MSU, Moscow 119234, Russia; (A.S.); (A.O.)
| | - Artem Fatkulin
- Laboratory of Molecular Physiology, HSE University, Moscow 101000, Russia
| | - Daria Surkova
- Department of Microbiology, Lomonosov MSU, Moscow 119234, Russia; (A.S.); (A.O.)
| | | | - Elizaveta Popova
- Department of Microbiology, Lomonosov MSU, Moscow 119234, Russia; (A.S.); (A.O.)
| |
Collapse
|
4
|
Gu Q, Mi L, Lai C, Guan X, Lu N, Zhan T, Wang G, Lu C, Xu L, Gao X, Zhang J. CPXM1 correlates to poor prognosis and immune cell infiltration in gastric cancer. Heliyon 2024; 10:e21909. [PMID: 38314284 PMCID: PMC10837494 DOI: 10.1016/j.heliyon.2023.e21909] [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: 12/12/2022] [Revised: 09/14/2023] [Accepted: 10/31/2023] [Indexed: 02/06/2024] Open
Abstract
Background Gastric cancer (GC) is the fourth most common cause of cancer-related death and the fifth most frequent malignant cancer, especially advanced GC. Carboxypeptidase X member 1 (CPXM1) is an epigenetic factor involved in many physiological processes, including osteoclast differentiation and adipogenesis. Several studies have shown the association of CPXM1 with multiple tumors; however, the mechanism of CPXM1 involvement in the progression of GC is yet to be characterized. Method CPXM1 expression data were obtained from the Tumor Immune Estimation Resource. The Cancer Genome Atlas and the Gene Expression Omnibus databases were used to obtain patient-matched clinicopathological information, and the Kaplan-Meier plot database was utilized for the prognosis analysis of GC patients. The Catalog of Somatic Mutations in Cancer and cBioportal databases were adopted to study CPXM1 mutations in tumors. Next, we utilized the Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene Set Enrichment Analysis for mechanism research. Furthermore, we performed tumor microenvironment and immune infiltration analysis based on CPXM1. Finally, we predicted sensitivity to several targeted drugs in GC patients based on CPXM1.CPXM1 is upregulated in GC and is correlated with poor prognosis, gender, and tumor stage in GC patients. Gene enrichment analysis suggested that CPXM1 may regulate the occurrence and progression of GC via the PI3K-AKT and TGF-β pathway. Moreover, CPXM1 expression results in an increase in the proportion of immune and stromal cells. Additionally, the proportion of plasma cells was inversely related to the expression of CPXM1, whereas macrophage M2 expression was proportionate to CPXM1 expression. Finally, six small-molecule drugs that showed notable variations in IC50 between two groups were screened. Conclusion These results suggested that CPXM1 regulates the progression of GC and may represent a novel target for the detection and treatment of GC.
Collapse
Affiliation(s)
- Qiou Gu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Lei Mi
- Department of Oncology, Yancheng First Hospital, Affilital Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, 66 Renmin South Road, Yancheng, Jiangsu, 210009, PR China
| | - Chuilin Lai
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Xiao Guan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Na Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Tian Zhan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Guoguang Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Chen Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Lei Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Xiang Gao
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| | - Jianping Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210009, PR China
| |
Collapse
|
5
|
Yamamoto T, Gi M, Yamashita S, Suzuki S, Fujioka M, Vachiraarunwong A, Guo R, Qiu G, Kakehashi A, Kato M, Uchida J, Wanibuchi H. DNA Methylation Aberrations in Dimethylarsinic Acid-Induced Bladder Carcinogenesis. Cancers (Basel) 2023; 15:5274. [PMID: 37958445 PMCID: PMC10648661 DOI: 10.3390/cancers15215274] [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: 10/02/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Arsenic is a known human urinary bladder carcinogen. While arsenic is known to cause aberrant DNA methylation, the mechanism of arsenic-triggered bladder carcinogenesis is not fully understood. The goal of this study was to identify aberrant DNA methylation in rat bladder urothelial carcinoma (UC) induced by dimethylarsinic acid (DMAV), a major organic metabolite of arsenic. We performed genome-wide DNA methylation and microarray gene expression analyses of DMAV-induced rat UCs and the urothelium of rats treated for 4 weeks with DMAV. We identified 40 genes that were both hypermethylated and downregulated in DMAV-induced rat UCs. Notably, four genes (CPXM1, OPCML, TBX20, and KCND3) also showed reduced expression in the bladder urothelium after 4 weeks of exposure to DMAV. We also found that CPXM1 is aberrantly methylated and downregulated in human bladder cancers and human bladder cancer cells. Genes with aberrant DNA methylation and downregulated expression in DMAV-exposed bladder urothelium and in DMAV-induced UCs in rats, suggest that these alterations occurred in the early stages of arsenic-induced bladder carcinogenesis. Further study to evaluate the functions of these genes will advance our understanding of the role of aberrant DNA methylation in arsenic bladder carcinogenesis, and will also facilitate the identification of new therapeutic targets for arsenic-related bladder cancers.
Collapse
Affiliation(s)
- Tomoki Yamamoto
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan; (T.Y.)
- Department of Molecular Urology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan
| | - Min Gi
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan; (T.Y.)
- Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan
| | - Satoshi Yamashita
- Department of Life Engineering, Faculty of Engineering, Maebashi Institute of Technology, 460-1 Kamisadori, Maebashi 371-0816, Gunma, Japan
| | - Shugo Suzuki
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan; (T.Y.)
| | - Masaki Fujioka
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan; (T.Y.)
| | - Arpamas Vachiraarunwong
- Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan
| | - Runjie Guo
- Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan
| | - Guiyu Qiu
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan; (T.Y.)
| | - Anna Kakehashi
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan; (T.Y.)
| | - Minoru Kato
- Department of Molecular Urology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan
| | - Junji Uchida
- Department of Molecular Urology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Osaka, Japan; (T.Y.)
| |
Collapse
|
6
|
Naschberger E, Flierl C, Huang J, Erkert L, Gamez-Belmonte R, Gonzalez-Acera M, Bober M, Mehnert M, Becker C, Schellerer VS, Britzen-Laurent N, Stürzl M. Analysis of the interferon-γ-induced secretome of intestinal endothelial cells: putative impact on epithelial barrier dysfunction in IBD. Front Cell Dev Biol 2023; 11:1213383. [PMID: 37645250 PMCID: PMC10460912 DOI: 10.3389/fcell.2023.1213383] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023] Open
Abstract
The development of inflammatory bowel diseases (IBD) involves the breakdown of two barriers: the epithelial barrier and the gut-vascular barrier (GVB). The destabilization of each barrier can promote initiation and progression of the disease. Interestingly, first evidence is available that both barriers are communicating through secreted factors that may accordingly serve as targets for therapeutic modulation of barrier functions. Interferon (IFN)-γ is among the major pathogenesis factors in IBD and can severely impair both barriers. In order to identify factors transmitting signals from the GVB to the epithelial cell barrier, we analyzed the secretome of IFN-γ-treated human intestinal endothelial cells (HIEC). To this goal, HIEC were isolated in high purity from normal colon tissues. HIEC were either untreated or stimulated with IFN-γ (10 U/mL). After 48 h, conditioned media (CM) were harvested and subjected to comparative hyper reaction monitoring mass spectrometry (HRM™ MS). In total, 1,084 human proteins were detected in the HIEC-CM. Among these, 43 proteins were present in significantly different concentrations between the CM of IFN-γ- and control-stimulated HIEC. Several of these proteins were also differentially expressed in various murine colitis models as compared to healthy animals supporting the relevance of these proteins secreted by inflammatory activated HIEC in the inter-barrier communication in IBD. The angiocrine pathogenic impact of these differentially secreted HIEC proteins on the epithelial cell barrier and their perspectives as targets to treat IBD by modulation of trans-barrier communication is discussed in detail.
Collapse
Affiliation(s)
- Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christian Flierl
- Division of Molecular and Experimental Surgery, Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jinghao Huang
- Division of Molecular and Experimental Surgery, Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lena Erkert
- Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Reyes Gamez-Belmonte
- Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Miguel Gonzalez-Acera
- Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | - Christoph Becker
- Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Vera S. Schellerer
- Department of Pediatric Surgery, University Medicine Greifswald, Greifswald, Germany
| | - Nathalie Britzen-Laurent
- Division of Surgical Research, Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
7
|
Fajardo D, Saint Jean R, Lyons PJ. Acquisition of new function through gene duplication in the metallocarboxypeptidase family. Sci Rep 2023; 13:2512. [PMID: 36781897 PMCID: PMC9925722 DOI: 10.1038/s41598-023-29800-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Gene duplication is a key first step in the process of expanding the functionality of a multigene family. In order to better understand the process of gene duplication and its role in the formation of new enzymes, we investigated recent duplication events in the M14 family of proteolytic enzymes. Within vertebrates, four of 23 M14 genes were frequently found in duplicate form. While AEBP1, CPXM1, and CPZ genes were duplicated once through a large-scale, likely whole-genome duplication event, the CPO gene underwent many duplication events within fish and Xenopus lineages. Bioinformatic analyses of enzyme specificity and conservation suggested a greater amount of neofunctionalization and purifying selection in CPO paralogs compared with other CPA/B enzymes. To examine the functional consequences of evolutionary changes on CPO paralogs, the four CPO paralogs from Xenopus tropicalis were expressed in Sf9 and HEK293T cells. Immunocytochemistry showed subcellular distribution of Xenopus CPO paralogs to be similar to that of human CPO. Upon activation with trypsin, the enzymes demonstrated differential activity against three substrates, suggesting an acquisition of new function following duplication and subsequent mutagenesis. Characteristics such as gene size and enzyme activation mechanisms are possible contributors to the evolutionary capacity of the CPO gene.
Collapse
Affiliation(s)
- Daniel Fajardo
- Department of Biology, Andrews University, Berrien Springs, MI, 49104, USA
| | - Ritchie Saint Jean
- Department of Biology, Andrews University, Berrien Springs, MI, 49104, USA
| | - Peter J Lyons
- Department of Biology, Andrews University, Berrien Springs, MI, 49104, USA.
| |
Collapse
|
8
|
Pervaz S, Ullah A, Adu-Gyamfi EA, Lamptey J, Sah SK, Wang MJ, Wang YX. Role of CPXM1 in Impaired Glucose Metabolism and Ovarian Dysfunction in Polycystic Ovary Syndrome. Reprod Sci 2023; 30:526-543. [PMID: 35697923 DOI: 10.1007/s43032-022-00987-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/20/2022] [Indexed: 11/24/2022]
Abstract
Polycystic ovary syndrome (PCOS), a common female endocrinopathy associated with both reproductive and metabolic disorders, has an unclear etiology and unsatisfactory management methods. Carboxypeptidase X, M14 family member 1 (CPXM1) is a protein involved in follicular atresia, insulin production, and adipose tissue production, though its role in PCOS is not fully understood. We used a 60% high-fat diet (HFD) plus dehydroepiandrosterone (DHEA)-induced PCOS mouse model to determine the role of CPXM1 in abnormal glucose metabolism and ovarian dysfunction in PCOS. We found that serum CPXM1 concentrations were higher in PCOS mice and positively correlated with increased levels of serum testosterone and insulin. In both ovarian and adipose tissues of PCOS mice, CPXM1 mRNA and protein levels were significantly increased but GLUT4 levels were significantly decreased. Immunohistochemistry (IHC) staining of the ovary showed increased CPXM1 expression in PCOS. In addition, the protein expression of phosphorylated protein kinase B (p-Akt) was also significantly decreased in PCOS mice. Furthermore, mRNA levels of inflammatory markers such as TNF-α, IL-6, IFN-α, and IFN-γ were increased in ovarian and adipose tissues of PCOS mice. However, IRS-1, IRS-2, and INSR levels were significantly decreased. Our results indicated for the first time that abnormally high expression of CPXM1, increased adiposity, impaired glucose tolerance, and chronic low-grade inflammation may act together in a vicious cycle in the pathophysiology of PCOS. Our research suggests the possibility of CPXM1 as a potential therapeutic target for the treatment of PCOS.
Collapse
Affiliation(s)
- Sadaf Pervaz
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, YiXueYuanLu Street No.1, YuZhong District, Chongqing, People's Republic of China
| | - Amin Ullah
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, YiXueYuanLu Street No.1, YuZhong District, Chongqing, People's Republic of China
| | - Enoch Appiah Adu-Gyamfi
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, YiXueYuanLu Street No.1, YuZhong District, Chongqing, People's Republic of China
| | - Jones Lamptey
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, YiXueYuanLu Street No.1, YuZhong District, Chongqing, People's Republic of China.,Department of Genetics, School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Sanjay Kumar Sah
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, YiXueYuanLu Street No.1, YuZhong District, Chongqing, People's Republic of China
| | - Mei-Jiao Wang
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, YiXueYuanLu Street No.1, YuZhong District, Chongqing, People's Republic of China. .,Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China.
| | - Ying-Xiong Wang
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, YiXueYuanLu Street No.1, YuZhong District, Chongqing, People's Republic of China. .,Department of Genetics, School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
9
|
Murase H, El-Sheikh Ali H, Ruby RE, Scoggin KE, Ball BA. Transcriptomic analysis of the chorioallantois in equine premature placental separation. Equine Vet J 2022; 55:405-418. [PMID: 35622344 DOI: 10.1111/evj.13602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 05/12/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Equine premature placental separation (PPS) is poorly understood and represents an important risk factor for fetal/neonatal hypoxia. OBJECTIVES To examine transcriptomic changes in the chorioallantois (CA) from mares with clinical PPS compared to the CA from normal foaling mares. Differential gene expression was determined and gene ontology as well as molecular pathways related to PPS were characterised. STUDY DESIGN Retrospective case: control study. METHODS CA were collected from Thoroughbred mares with a clinical history of PPS (n=33) and from control Thoroughbred mares (n=4) with normal parturition for examination of transcriptional changes in the placenta associated with PPS. Transcriptomic changes in the villous CA near the cervical star were determined by Illumina® sequencing and subsequent bioinformatic analysis. PPS samples were divided by k-means clustering, and differentially expressed genes (DEGs) in each PPS cluster were identified by comparing to controls. Shared DEGs between PPS clusters were used for gene ontology analysis and pathway analysis. RESULTS A total of 1204 DEGs were identified between PPS and control. Gene ontology revealed extracellular matrix (ECM) and cell adhesion, and pathway analysis revealed fatty acid, p-53, hypoxia, and inflammation. Eleven key regulator genes of PPS including growth factors (IGF1, TGFB2, TGFB3), transcription factors (HIF1A, JUNB, SMAD3), and transmembrane receptors (FGFR1, TNFRSF1A, TYROBP) were also identified. MAIN LIMITATIONS The use of clinical history of PPS, in the absence of other criteria, may have led to misidentification of some cases as PPS. CONCLUSIONS Transcriptomic analysis indicated that changes in ECM and cell adhesion were important factors in equine PPS. Key predicted upstream events include genes associated with hypoxia, inflammation and growth factors related to the pathogenesis of equine PPS.
Collapse
Affiliation(s)
- Harutaka Murase
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA.,Equine Science Division, Hidaka Training and Research Center, Japan Racing Association, Urakawa, Hokkaido, Japan
| | - Hossam El-Sheikh Ali
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA.,Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Rebecca E Ruby
- Veterinary Diagnostic Laboratory, University of Kentucky, Lexington, KY, USA
| | - Kirsten E Scoggin
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Barry A Ball
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
10
|
MiRNA-29b and miRNA-497 Modulate the Expression of Carboxypeptidase X Member 2, a Candidate Gene Associated with Left Ventricular Hypertrophy. Int J Mol Sci 2022; 23:ijms23042263. [PMID: 35216380 PMCID: PMC8880112 DOI: 10.3390/ijms23042263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 01/27/2023] Open
Abstract
Left ventricular hypertrophy (LVH) is a major risk factor for adverse cardiovascular events. Recently, a novel candidate gene encoding the carboxypeptidase X member 2 (CPXM2) was found to be associated with hypertension-induced LVH. CPXM2 belongs to the M14 family of metallocarboxypeptidases, yet it lacks detectable enzyme activity, and its function remains unknown. Here, we investigated the impact of micro (mi)RNA-29b, miRNA-195, and miRNA-497 on the posttranscriptional expression control of CPXM2. Candidate miRNAs for CPXM2 expression control were identified in silico. CPXM2 expression in rat cardiomyocytes (H9C2) was characterized via real-time PCR, Western blotting, and immunofluorescence. Direct miRNA/target mRNA interaction was analysed by dual luciferase assay. CPXM2 was expressed in H9C2 and co-localised with z-disc associated protein PDZ and LIM domain 3 (Pdlim3). Transfection of H9C2 with miRNA-29b, miRNA-195, and miRNA-497 led to decreased levels of CPXM2 mRNA and protein, respectively. Results of dual luciferase assays revealed that miRNA-29b and miRNA-497, but not miRNA-195, directly regulated CPXM2 expression on a posttranscriptional level via binding to the 3′UTR of CPXM2 mRNA. We identified two miRNAs capable of the direct posttranscriptional expression control of CPXM2 expression in rat cardiomyocytes. This novel data may help to shed more light on the—so far—widely unexplored expression control of CPXM2 and its potential role in LVH.
Collapse
|
11
|
Goto K, Osaki M, Izutsu R, Tanaka H, Sasaki R, Tanio A, Satofuka H, Kazuki Y, Yamamoto M, Kugoh H, Ito H, Oshimura M, Fujiwara Y, Okada F. Establishment of an antibody specific for AMIGO2 improves immunohistochemical evaluation of liver metastases and clinical outcomes in patients with colorectal cancer. Diagn Pathol 2022; 17:16. [PMID: 35094710 PMCID: PMC8802484 DOI: 10.1186/s13000-021-01176-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/26/2021] [Indexed: 12/11/2022] Open
Abstract
Abstract
Instruction
The human amphoterin-induced gene and open reading frame (AMIGO) was identified as a novel cell adhesion molecule of type I transmembrane protein. AMIGO2 is one of three members of the AMIGO family (AMIGO1, 2, and 3), and the similarity between them is approximately 40% at the amino acid level. We have previously shown that AMIGO2 functions as a driver of liver metastasis. Immunohistochemical analysis of AMIGO2 expression in colorectal cancer (CRC) using a commercially available anti-AMIGO2 mouse monoclonal antibody clone sc-373699 (sc mAb) correlated with liver metastasis and poor prognosis. However, the sc mAb was found to be cross-reactive with all three molecules in the AMIGO family.
Methods
We generated a rat monoclonal antibody clone rTNK1A0012 (rTNK mAb) for human AMIGO2. The rTNK mAb was used to re-evaluate the association between AMIGO2 expression and liver metastases/clinical outcomes using the same CRC tissue samples previously reported with sc mAb.
Results
Western blot analysis revealed that a rTNK mAb was identified as being specific for AMIGO2 protein and did not cross-react with AMIGO1 and AMIGO3. The rTNK mAb and sc mAb showed higher AMIGO2 expression, which correlates with a high frequency of liver metastases (65.3% and 47.5%, respectively), while multivariate analysis showed that AMIGO2 expression was an independent prognostic factor for liver metastases (p = 7.930E-10 and p = 1.707E-5). The Kaplan-Meier analyses showed that the rTNK mAb (p = 0.004), but not sc mAb (p = 0.107), predicted worse overall survival in patients with high AMIGO2 expression. The relationship between AMIGO2 expression and poor disease-specific survival showed a higher level of significance for rTNK mAb (p = 0.00004) compared to sc mAb (p = 0.001).
Conclusions
These results indicate that the developed rTNK1A0012 mAb is an antibody that specifically recognizes AMIGO2 by immunohistochemistry and can be a more reliable and applicable method for the diagnostic detection of liver metastases and worse prognosis in patients with high AMIGO2-expressing CRC.
Collapse
|
12
|
Mining for encrypted peptide antibiotics in the human proteome. Nat Biomed Eng 2021; 6:67-75. [PMID: 34737399 DOI: 10.1038/s41551-021-00801-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 08/25/2021] [Indexed: 12/30/2022]
Abstract
The emergence of drug-resistant bacteria calls for the discovery of new antibiotics. Yet, for decades, traditional discovery strategies have not yielded new classes of antimicrobial. Here, by mining the human proteome via an algorithm that relies on the sequence length, net charge, average hydrophobicity and other physicochemical properties of antimicrobial peptides, we report the identification of 2,603 encrypted peptide antibiotics that are encoded in proteins with biological function unrelated to the immune system. We show that the encrypted peptides kill pathogenic bacteria by targeting their membrane, modulate gut and skin commensals, do not readily select for bacterial resistance, and possess anti-infective activity in skin abscess and thigh infection mouse models. We also show, in vitro and in the two mouse models of infection, that encrypted antibiotic peptides from the same biogeographical area display synergistic antimicrobial activity. Our algorithmic strategy allows for the rapid mining of proteomic data and opens up new routes for the discovery of candidate antibiotics.
Collapse
|
13
|
McDonald RC, Schott MJ, Idowu TA, Lyons PJ. Biochemical and genetic analysis of Ecm14, a conserved fungal pseudopeptidase. BMC Mol Cell Biol 2020; 21:86. [PMID: 33256608 PMCID: PMC7706225 DOI: 10.1186/s12860-020-00330-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/18/2020] [Indexed: 01/28/2023] Open
Abstract
Background Like most major enzyme families, the M14 family of metallocarboxypeptidases (MCPs) contains a number of pseudoenzymes predicted to lack enzyme activity and with poorly characterized molecular function. The genome of the yeast Saccharomyces cerevisiae encodes one member of the M14 MCP family, a pseudoenzyme named Ecm14 proposed to function in the extracellular matrix. In order to better understand the function of such pseudoenzymes, we studied the structure and function of Ecm14 in S. cerevisiae. Results A phylogenetic analysis of Ecm14 in fungi found it to be conserved throughout the ascomycete phylum, with a group of related pseudoenzymes found in basidiomycetes. To investigate the structure and function of this conserved protein, His6-tagged Ecm14 was overexpressed in Sf9 cells and purified. The prodomain of Ecm14 was cleaved in vivo and in vitro by endopeptidases, suggesting an activation mechanism; however, no activity was detectable using standard carboxypeptidase substrates. In order to determine the function of Ecm14 using an unbiased screen, we undertook a synthetic lethal assay. Upon screening approximately 27,000 yeast colonies, twenty-two putative synthetic lethal clones were identified. Further analysis showed many to be synthetic lethal with auxotrophic marker genes and requiring multiple mutations, suggesting that there are few, if any, single S. cerevisiae genes that present synthetic lethal interactions with ecm14Δ. Conclusions We show in this study that Ecm14, although lacking detectable enzyme activity, is a conserved carboxypeptidase-like protein that is secreted from cells and is processed to a mature form by the action of an endopeptidase. Our study and datasets from other recent large-scale screens suggest a role for Ecm14 in processes such as vesicle-mediated transport and aggregate invasion, a fungal process that has been selected against in modern laboratory strains of S. cerevisiae. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-020-00330-w.
Collapse
Affiliation(s)
| | - Matthew J Schott
- Department of Biology, Andrews University, Berrien Springs, MI, USA
| | - Temitope A Idowu
- Department of Biology, Andrews University, Berrien Springs, MI, USA
| | - Peter J Lyons
- Department of Biology, Andrews University, Berrien Springs, MI, USA.
| |
Collapse
|
14
|
Wang YH, Lin CC, Yao CY, Hsu CL, Hou HA, Tsai CH, Chou WC, Tien HF. A 4-gene leukemic stem cell score can independently predict the prognosis of myelodysplastic syndrome patients. Blood Adv 2020; 4:644-654. [PMID: 32078680 PMCID: PMC7042996 DOI: 10.1182/bloodadvances.2019001185] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/24/2020] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndrome (MDS) comprised a heterogeneous group of diseases. The prognosis of patients varies even in the same risk groups. Searching for novel prognostic markers is warranted. Leukemic stem cells (LSCs) are responsible for chemoresistance and relapse in leukemia. Recently, expressions of 17 genes related to stemness of LSCs were found to be associated with prognosis in acute myeloid leukemia patients. However, the clinical impact of LSC genes expressions in MDS, a disorder arising from hematopoietic stem cells, remains unclear. We analyzed expression profile of the 17 stemness-related genes in primary MDS patients and identified expression of 4 genes (LAPTM4B, NGFRAP1, EMP1, and CPXM1) were significantly correlated with overall survival (OS). We constructed an LSC4 scoring system based on the weighted sums of the expression of 4 genes and explored its clinical implications in MDS patients. Higher LSC4 scores were associated with higher revised International Prognostic Scoring System (IPSS-R) scores, complex cytogenetics, and mutations in RUNX1, ASXL1, and TP53. High-score patients had significantly shorter OS and leukemia-free survival (LFS), which was also confirmed in 2 independent validation cohorts. Subgroup analysis revealed the prognostic significance of LSC4 scores for OS remained valid across IPSS-R lower- and higher-risk groups. Furthermore, higher LSC4 score was an independent adverse risk factor for OS and LFS in multivariate analysis. In summary, LSC4 score can independently predict prognosis in MDS patients irrespective of IPSS-R risks and may be used to guide the treatment of MDS patients, especially lower-risk group in whom usually only supportive treatment is given.
Collapse
Affiliation(s)
- Yu-Hung Wang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; and
- Division of Hematology, Department of Internal Medicine
| | - Chien-Chin Lin
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; and
- Division of Hematology, Department of Internal Medicine
- Department of Laboratory Medicine, and
| | - Chi-Yuan Yao
- Division of Hematology, Department of Internal Medicine
- Department of Laboratory Medicine, and
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine
| | | | - Wen-Chien Chou
- Division of Hematology, Department of Internal Medicine
- Department of Laboratory Medicine, and
| | | |
Collapse
|
15
|
Kumar A, Bandapalli OR, Paramasivam N, Giangiobbe S, Diquigiovanni C, Bonora E, Eils R, Schlesner M, Hemminki K, Försti A. Familial Cancer Variant Prioritization Pipeline version 2 (FCVPPv2) applied to a papillary thyroid cancer family. Sci Rep 2018; 8:11635. [PMID: 30072699 PMCID: PMC6072708 DOI: 10.1038/s41598-018-29952-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022] Open
Abstract
Whole-genome sequencing methods in familial cancer are useful to unravel rare clinically important cancer predisposing variants. Here, we present improvements in our pedigree-based familial cancer variant prioritization pipeline referred as FCVPPv2, including 12 tools for evaluating deleteriousness and 5 intolerance scores for missense variants. This pipeline is also capable of assessing non-coding regions by combining FANTOM5 data with sets of tools like Bedtools, ChromHMM, Miranda, SNPnexus and Targetscan. We tested this pipeline in a family with history of a papillary thyroid cancer. Only one variant causing an amino acid change G573R (dbSNP ID rs145736623, NM_019609.4:exon11:c.G1717A:p.G573R) in the carboxypeptidase gene CPXM1 survived our pipeline. This variant is located in a highly conserved region across vertebrates in the peptidase_M14 domain (Pfam ID PF00246). The CPXM1 gene may be involved in adipogenesis and extracellular matrix remodelling and it has been suggested to be a tumour suppressor in breast cancer. However, the presence of the variant in the ExAC database suggests it to be a rare polymorphism or a low-penetrance risk allele. Overall, our pipeline is a comprehensive approach for prediction of predisposing variants for high-risk cancer families, for which a functional characterization is a crucial step to confirm their role in cancer predisposition.
Collapse
Affiliation(s)
- Abhishek Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany.
| | - Obul Reddy Bandapalli
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany.
| | - Nagarajan Paramasivam
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, D69120, Heidelberg, Germany
| | - Sara Giangiobbe
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany
| | | | - Elena Bonora
- Unit of Medical Genetics, S.Orsola-Malpighi Hospital, 40138, Bologna, Italy
| | - Roland Eils
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology (IPMB) and BioQuant, Heidelberg University, D69120, Heidelberg, Germany
| | - Matthias Schlesner
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), D69120, Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| |
Collapse
|
16
|
Kim YH, Barclay JL, He J, Luo X, O'Neill HM, Keshvari S, Webster JA, Ng C, Hutley LJ, Prins JB, Whitehead JP. Identification of carboxypeptidase X (CPX)-1 as a positive regulator of adipogenesis. FASEB J 2016; 30:2528-40. [PMID: 27006448 DOI: 10.1096/fj.201500107r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/10/2016] [Indexed: 01/13/2023]
Abstract
Adipose tissue expansion occurs through a combination of hypertrophy of existing adipocytes and generation of new adipocytes via the process of hyperplasia, which involves the proliferation and subsequent differentiation of preadipocytes. Deficiencies in hyperplasia contribute to adipose tissue dysfunction and the association of obesity with chronic cardiometabolic diseases. Thus, increased understanding of hyperplastic pathways may be expected to afford novel therapeutic strategies. We have reported that fibroblast growth factor (FGF)-1 promotes proliferation and differentiation of human preadipocytes and recently demonstrated that bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) is a central, proximal effector. Herein, we describe the identification and characterization of carboxypeptidase X (CPX)-1, a secreted collagen-binding glycoprotein, as a novel downstream effector in human primary and Simpson-Golabi-Behmel syndrome preadipocytes. CPX-1 expression increased after treatment of preadipocytes with FGF-1, BAMBI knockdown, or induction of differentiation. CPX-1 knockdown compromised preadipocyte differentiation coincident with reduced collagen expression. Furthermore, preadipocytes differentiated on matrix derived from CPX-1 knockdown cells exhibited reduced Glut4 expression and insulin-stimulated glucose uptake. Finally, CPX-1 expression was increased in adipose tissue from obese mice and humans. Collectively, these findings establish CPX-1 as a positive regulator of adipogenesis situated downstream of FGF-1/BAMBI that may contribute to hyperplastic adipose tissue expansion via affecting extracellular matrix remodeling.-Kim, Y.-H., Barclay, J. L., He, J., Luo, X., O'Neill, H. M., Keshvari, S., Webster, J. A., Ng, C., Hutley, L. J., Prins, J. B., Whitehead, J. P. Identification of carboxypeptidase X (CPX)-1 as a positive regulator of adipogenesis.
Collapse
Affiliation(s)
- Yu-Hee Kim
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Johanna L Barclay
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Jingjing He
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Xiao Luo
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Hayley M O'Neill
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Sahar Keshvari
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Julie A Webster
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Choaping Ng
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Louise J Hutley
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Johannes B Prins
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Jonathan P Whitehead
- Metabolic Medicine Group, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|