1
|
Lei P, Yu H, Jiang T, Ma J, Du J, Fang Y, Wang H, Chen R, Yang Q, Cheng Y, Wu W, Sun D. Development of a sodium hyaluronate-enriched therapeutic formulation with stevia glycoside and mogroside V for the comprehensive management of diabetes and its complications. Int J Biol Macromol 2025; 293:139487. [PMID: 39756763 DOI: 10.1016/j.ijbiomac.2025.139487] [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: 10/13/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Diabetes prevalence continues to increase as a result of people's increasing sugar intake. Diabetes mellitus and its complications (dry skin, constipation, depression, and dental caries), as well as the prohibition of sweets ingestion, seriously affect patients' physical and mental health. Therefore, it is crucial to develop a long-term food for special medical purposes (FSMP) that aids in managing diabetes and its complications. To ensure effective biomedical function and taste, we developed a FSMP beverage formulation containing stevia glycoside, mogroside V, and sodium hyaluronate (SMH-B), each at a concentration of 0.1 mg/mL. Meanwhile, this study verified that SMH-B is an environmentally friendly and biocompatible formulation. Furthermore, both in vivo and in vitro studies have demonstrated that SMH-B significantly lowers blood glucose and lipid levels, enhances skin moisture and elasticity, prevents dental caries, alleviates constipation, reduces oxidative stress, and mitigates depressive symptoms. Notably, the SMH-B compound formula exhibits a more effective adjuvant therapeutic effect compared to single-ingredient formulation composed of stevia glycosides, mogroside V, and sodium hyaluronate. Moreover, SMH-B provides the sweetness desired by diabetic patients without affecting blood glucose levels, while also offering an auxiliary therapeutic role, making it a potential FSMP for diabetes management.
Collapse
Affiliation(s)
- Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiao Du
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, 999077, Hong Kong
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Hong Kong
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Yongwei Cheng
- National Engineering Research Center of Cell Growth Factor Drugs and Protein Biologics, Wenzhou Medical University, Wenzhou 325000, China; MedTech (Wenzhou) Health Innovation Achievement Transformation Institute, Wenzhou Institute of Industry & Science, Wenzhou 325000, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
2
|
Khan N, Li Z, Ali A, Quan B, Kang J, Ullah M, Yin XJ, Shafiq M. Comprehensive transcriptomic analysis of myostatin-knockout pigs: insights into muscle growth and lipid metabolism. Transgenic Res 2025; 34:12. [PMID: 39979478 DOI: 10.1007/s11248-025-00431-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
Pigs are a vital source of protein worldwide, contributing approximately 43% of global meat production. Recent genetic advancements in the myostatin (MSTN) gene have facilitated the development of double-muscling traits in livestock. In this study, we investigate the transcriptomic profiles of second-generation MSTN-knockout (MSTN-/-) pigs, generated through CRISPR/Cas9 gene editing and somatic cell nuclear transfer (SCNT). Using RNA sequencing, we compared the transcriptomic landscapes of muscle tissues from MSTN-/- pigs and wild-type (WT) counterparts. The sequencing yielded an average unique read mapping rate of 86.7% to the Sus scrofa reference genome. Our analysis revealed 15,142 differentially expressed genes (DEGs), including 121 novel genes, with 2554 genes upregulated and 1629 downregulated in the MSTN-/- group relative to the wild-type group. Notable transcriptomic changes were identified in genes associated with muscle development, lipid metabolism, and other physiological processes. These findings provide valuable insights into the molecular consequences of MSTN inactivation, with potential applications in the optimization of livestock breeding and advancements in biomedical research.
Collapse
Affiliation(s)
- Nasar Khan
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Department of Animal Science, Yanbian University, Yanji, Jilin, 133002, China
| | - Zhouyan Li
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Department of Animal Science, Yanbian University, Yanji, Jilin, 133002, China
| | - Akbar Ali
- School of Life Sciences, Liaoning University, Shenyang, 110036, China
| | - Biaohu Quan
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Department of Animal Science, Yanbian University, Yanji, Jilin, 133002, China
| | - Jindan Kang
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Department of Animal Science, Yanbian University, Yanji, Jilin, 133002, China
| | - Munib Ullah
- Department of Clinical Studies, Faculty of Veterinary and Animal Sciences, Pir Mehr Ali Shah Arid Agriculture University, Rawalpindi, 46000, Pakistan
| | - Xi-Jun Yin
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Department of Animal Science, Yanbian University, Yanji, Jilin, 133002, China.
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
3
|
Chen B, Li S, Cheng X, Geng W, Cheng Z, Zhang K, Ma C, Challioui MK, Aayadi SE, Guo Y, Wang Y, Wang D, Tian W, Li H, Liu X. Functional analysis of key members affecting egg production in the transglutaminase gene family in chickens. Poult Sci 2025; 104:104794. [PMID: 39813865 PMCID: PMC11782901 DOI: 10.1016/j.psj.2025.104794] [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: 10/25/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
The transglutaminase (TGMs) family plays a crucial role in regulating mammalian reproduction, yet its impact on poultry reproductive traits has not been extensively studied. In this study, we identified eight members of the TGMs family in chickens and examined the contributions of genetic variations of coagulation factor XIII A chain (F13A1), transglutaminase 4 (TGM4), and LOC101749664 to selective breeding in commercial layers through genetic variation response pattern analysis. Transcriptome data from various tissues of high- and low-egg-yielding Gushi chickens revealed significant positive correlations between the mRNA expression levels of TGM4 and F13A1 genes and egg production (P < 0.05). Genotype-phenotype correlation analysis showed that 26 single nucleotide polymorphisms (SNPs) within genomic sequences of TGM4 and F13A1 genes exhibited significant associations with egg production traits across different laying periods. Additionally, spatiotemporal expression patterns of TGM4 in ovarian follicles between high- and low-laying chicken breeds determined by RT-qPCR highlighted the pivotal role of TGM4 in follicle development. Functionally, the gain-of-function assay demonstrated that TGM4 promoted granulosa cell proliferation, inhibited apoptosis, and enhanced progesterone synthesis. In conclusion, this is the first study to elucidate the role of the TGMs gene family in regulating egg production in chickens, offering new insights for the selective breeding of layers with improved egg-laying traits.
Collapse
Affiliation(s)
- Botong Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Shuohan Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Xi Cheng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Wanzhuo Geng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zhimin Cheng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Ke Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Chenglin Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Mohammed Kamal Challioui
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Department of Animal Production and Biotechnology, Institut Agronomique et Vétérinaire Hassan II, BP, 6202, Rabat, Morocco
| | - Soufiane El Aayadi
- Department of Animal Production and Biotechnology, Institut Agronomique et Vétérinaire Hassan II, BP, 6202, Rabat, Morocco
| | - Yulong Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yangyang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Dandan Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, 453003, China
| | - Weihua Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, 450046, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, 450046, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, 450046, China.
| |
Collapse
|
4
|
Soltani F, Welman M, Samani SE, Pacis A, Lordkipanidzé M, Kaartinen MT. Factor XIII-A Transglutaminase Contributes to Neutrophil Extracellular Trap (NET)-mediated Fibrin(ogen) Network Formation and Crosslinking. Thromb Haemost 2025. [PMID: 39694058 DOI: 10.1055/a-2504-1559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
BACKGROUND Neutrophil extracellular traps can contribute to thrombosis via stabilization of fibrin network, which is normally conducted by plasma transglutaminase, Factor XIII-A as part of coagulation cascade. The possible presence and activity of FXIII-A in neutrophils or during NETosis are unknown. Here, we investigated potential presence of FXIII-A in neutrophils and participation in NET-fibrin(ogen) interaction in vitro. METHODS: Data mining of human and mouse F13A1/F13a1 mRNA expression in whole-body scRNA sequence atlases was conducted. F13a1 mRNA and protein expression was assessed in isolated mouse bone marrow neutrophils. NETosis was induced using 12-phorbol 13-myristate acetate (PMA), and the transglutaminase activity was assessed with 5-(biotinamido)pentylamine incorporation to plasma fibronectin and a fluorescence-fibrin(ogen)-based activity assay using ATTO488-Cadaverine. Externalization of FXIII-A and its interaction with neutrophil extracellular trap (NET) markers, namely, decondensed DNA, CitH3, and MPO, were examined with immunofluorescence microscopy. NET-fibrin(ogen) interaction was investigated with and without serum and/or transglutaminase inhibitor, NC9. Effect of soluble fibrinogen and fibrin(ogen) network on NETosis was also assessed. RESULTS Data mining of RNAseq atlases showed F13A1/F13a1 expression in adipose tissue, blood, and bone marrow neutrophils. mRNA expression and protein production were confirmed in isolated neutrophils where expression was comparable to that of macrophages and monocytes. FXIII-A was externalized and active as a transglutaminase and colocalized with NET markers during NETosis. FXIII-A transglutaminase activity promoted NET-fibrin(ogen) interaction and entrapment of neutrophils within fibrin(ogen) matrix. Soluble fibrinogen or fibrin(ogen) network did not induce NETosis. CONCLUSION This study identifies neutrophils as a source of FXIII-A and suggests its role in stabilizing NET-fibrin(ogen) matrix structures.
Collapse
Affiliation(s)
- Fatemeh Soltani
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Mélanie Welman
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Sahar Ebrahimi Samani
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Alain Pacis
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
- Canadian Centre for Computational Genomics, McGill University, Montreal, Quebec, Canada
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculté de Pharmacie, Université de Montréal, Montreal, Quebec, Canada
| | - Mari T Kaartinen
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| |
Collapse
|
5
|
Green CR, Alaeddine LM, Wessendorf-Rodriguez KA, Turner R, Elmastas M, Hover JD, Murphy AN, Ryden M, Mejhert N, Metallo CM, Wallace M. Impaired branched-chain amino acid (BCAA) catabolism during adipocyte differentiation decreases glycolytic flux. J Biol Chem 2024; 300:108004. [PMID: 39551140 DOI: 10.1016/j.jbc.2024.108004] [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: 08/15/2023] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024] Open
Abstract
Dysregulated branched-chain amino acid (BCAA) metabolism has emerged as a key metabolic feature associated with the obese insulin-resistant state, and adipose BCAA catabolism is decreased in this context. BCAA catabolism is upregulated early in adipogenesis, but the impact of suppressing this pathway on the broader metabolic functions of the resultant adipocyte remains unclear. Here, we use CRISPR/Cas9 to decrease BCKDHA in 3T3-L1 and human pre-adipocytes, and ACAD8 in 3T3-L1 pre-adipocytes to induce a deficiency in BCAA catabolism through differentiation. We characterize the transcriptional and metabolic phenotype of 3T1-L1 cells using RNAseq and 13C metabolic flux analysis within a network spanning glycolysis, tricarboxylic acid (TCA) metabolism, BCAA catabolism, and fatty acid synthesis. While lipid droplet accumulation is maintained in Bckdha-deficient adipocytes, they display a more fibroblast-like transcriptional signature. In contrast, Acad8 deficiency minimally impacts gene expression. Decreased glycolytic flux emerges as the most distinct metabolic feature of 3T3-L1 Bckdha-deficient cells, accompanied by a ∼40% decrease in lactate secretion, yet pyruvate oxidation and utilization for de novo lipogenesis is increased to compensate for the loss of BCAA carbon. Deletion of BCKDHA in human adipocyte progenitors also led to a decrease in glucose uptake and lactate secretion; however, these cells did not upregulate pyruvate utilization, and lipid droplet accumulation and expression of adipocyte differentiation markers was decreased in BCKDH knockout cells. Overall our data suggest that human adipocyte differentiation may be more sensitive to the impact of decreased BCKDH activity than 3T3-L1 cells and that both metabolic and regulatory cross-talk exist between BCAA catabolism and glycolysis in adipocytes. Suppression of BCAA catabolism associated with metabolic syndrome may result in a metabolically compromised adipocyte.
Collapse
Affiliation(s)
- Courtney R Green
- Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA; Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Lynn M Alaeddine
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Karl A Wessendorf-Rodriguez
- Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA; Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Rory Turner
- School of Agriculture and Food Science, University College Dublin, Belfield, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| | - Merve Elmastas
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Justin D Hover
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Anne N Murphy
- Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Mikael Ryden
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Niklas Mejhert
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Christian M Metallo
- Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA; Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Martina Wallace
- School of Agriculture and Food Science, University College Dublin, Belfield, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland.
| |
Collapse
|
6
|
Hernaiz A, Marín B, Vázquez FJ, Badiola JJ, Zaragoza P, Bolea R, Martín-Burriel I. RNA-sequencing transcriptomic analysis of scrapie-exposed ovine mesenchymal stem cells. Res Vet Sci 2024; 180:105423. [PMID: 39341025 DOI: 10.1016/j.rvsc.2024.105423] [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: 05/21/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
In neurodegenerative diseases, including prion diseases, cellular models arise as useful tools to study the pathogenic mechanisms occurring in these diseases and to assess the efficacy of potential therapeutic compounds. In the present study, a RNA-sequencing analysis of bone marrow-derived ovine mesenchymal stem cells (oBM-MSCs) exposed to scrapie brain homogenate was performed to try to unravel genes and pathways potentially involved in prion diseases and MSC response mechanisms to prions. The oBM-MSCs were cultured in three different conditions (inoculated with brain homogenate of scrapie-infected sheep, with brain homogenate of healthy sheep and in standard growth conditions without inoculum) that were analysed at two exposure times: 2 and 4 days post-inoculation (dpi). Differentially expressed genes (DEGs) in scrapie-treated oBM-MSCs were found in the two exposure times finding the higher number at 2 dpi, which coincided with the inoculum removal time. Pathways enriched in DEGs were related to biological functions involved in prion toxicity and MSC response to the inflammatory environment of scrapie brain homogenate. Moreover, RNA-sequencing analysis was validated amplifying by RT-qPCR a set of 11 DEGs with functions related with prion propagation and its associated toxicity. Seven of these genes displayed significant expression changes in scrapie-treated cells. These results contribute to the knowledge of the molecular mechanisms behind the early toxicity observed in these cells after prion exposure and to elucidate the response of MSCs to neuroinflammation.
Collapse
Affiliation(s)
- Adelaida Hernaiz
- Laboratorio de Genética Bioquímica (LAGENBIO), Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain; Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2) UNIZAR-CITA, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS-Aragón), Zaragoza, Spain
| | - Belén Marín
- Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2) UNIZAR-CITA, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS-Aragón), Zaragoza, Spain; Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Francisco J Vázquez
- Laboratorio de Genética Bioquímica (LAGENBIO), Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain; Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2) UNIZAR-CITA, Zaragoza, Spain; Departamento de Patología Animal, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Juan J Badiola
- Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2) UNIZAR-CITA, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS-Aragón), Zaragoza, Spain; Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica (LAGENBIO), Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain; Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2) UNIZAR-CITA, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS-Aragón), Zaragoza, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Bolea
- Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2) UNIZAR-CITA, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS-Aragón), Zaragoza, Spain; Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Inmaculada Martín-Burriel
- Laboratorio de Genética Bioquímica (LAGENBIO), Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain; Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2) UNIZAR-CITA, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS-Aragón), Zaragoza, Spain; Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
7
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
8
|
Peng M, Zou R, Yao S, Meng X, Wu W, Zeng F, Chen Z, Yuan S, Zhao F, Liu W. High-intensity interval training and medium-intensity continuous training may affect cognitive function through regulation of intestinal microbial composition and its metabolite LPS by the gut-brain axis. Life Sci 2024; 352:122871. [PMID: 38936602 DOI: 10.1016/j.lfs.2024.122871] [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: 03/01/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024]
Abstract
AIMS The gut-brain axis is the communication mechanism between the gut and the central nervous system, and the intestinal flora and lipopolysaccharide (LPS) play a crucial role in this mechanism. Exercise regulates the gut microbiota composition and metabolite production (i.e., LPS). We aimed to investigate the effects of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on cognitive function in C57BL/6 J mice through gut-brain axis regulation of gut microbiota composition and LPS displacement. MAIN METHODS C57BL/6 J male mice were randomly divided into sedentary, HIIT, and MICT groups. After 12 weeks of exercise intervention, the cognitive function of the brain and mRNA levels of related inflammatory factors were measured. RNA sequencing, Golgi staining, intestinal microbial 16 s rDNA sequencing, and ELISA were performed. KEY FINDINGS HIIT and MICT affect brain cognitive function by regulating the gut microbiota composition and its metabolite, LPS, through the gut microbiota-gut-brain axis. HIIT is suspected to have a risk: it can induce "intestinal leakage" by regulating intestinal permeability-related microbiota, resulting in excessive LPS in the blood and brain and activating M1 microglia in the brain, leading to reduced dendritic spine density and affecting cognitive function. SIGNIFICANCE This study revealed a potential link between changes in the gut microbiota and cognitive function. It highlighted the possible risk of HIIT in reducing dendritic spine density and affecting cognitive function.
Collapse
Affiliation(s)
- Mei Peng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Ruihan Zou
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Sisi Yao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Xiangyuan Meng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Shunling Yuan
- Yangtze University College of Arts and Sciences, Jingzhou 434020, China
| | - Fei Zhao
- The First Affiliated Hospital of Hunan Normal University, Changsha 410002, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China; Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
9
|
Mahmoodi M, Mirzarazi Dahagi E, Nabavi M, Penalva YCM, Gosaine A, Murshed M, Couldwell S, Munter LM, Kaartinen MT. Circulating plasma fibronectin affects tissue insulin sensitivity, adipocyte differentiation, and transcriptional landscape of adipose tissue in mice. Physiol Rep 2024; 12:e16152. [PMID: 39054559 PMCID: PMC11272447 DOI: 10.14814/phy2.16152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
Plasma fibronectin (pFN) is a hepatocyte-derived circulating extracellular matrix protein that affects cell morphology, adipogenesis, and insulin signaling of adipocytes in vitro. In this study, we show pFN accrual to adipose tissue and its contribution to tissue homeostasis in mice. Hepatocyte-specific conditional Fn1 knockout mice (Fn1-/-ALB) show a decrease in adipose tissue FN levels and enhanced insulin sensitivity of subcutaneous (inguinal), visceral (epididymal) adipose tissue on a normal diet. Diet-induced obesity model of the Fn1-/-ALB mouse showed normal weight gain and whole-body fat mass, and normal adipose tissue depot volumes and unaltered circulating leptin and adiponectin levels. However, Fn1-/-ALB adipose depots showed significant alterations in adipocyte size and gene expression profiles. The inguinal adipose tissue on a normal diet, which had alterations in fatty acid metabolism and thermogenesis suggesting browning. The presence of increased beige adipocyte markers Ucp1 and Prdm16 supported this. In the inguinal fat, the obesogenic diet resulted in downregulation of the browning markers and changes in gene expression reflecting development, morphogenesis, and mesenchymal stem cell maintenance. Epididymal adipose tissue showed alterations in developmental and stem cell gene expression on both diets. The data suggests a role for pFN in adipose tissue insulin sensitivity and cell profiles.
Collapse
Affiliation(s)
- Mahdokht Mahmoodi
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
| | - Elahe Mirzarazi Dahagi
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Mir‐Hamed Nabavi
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
| | - Ylauna C. M. Penalva
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Centre de Recherche en Biologie Structurale (CRBS)McGill UniversityMontrealQuebecCanada
| | - Amrita Gosaine
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Monzur Murshed
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
- Shriners Hospital for ChildrenMontrealQuebecCanada
| | - Sandrine Couldwell
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
| | - Lisa M. Munter
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Centre de Recherche en Biologie Structurale (CRBS)McGill UniversityMontrealQuebecCanada
| | - Mari T. Kaartinen
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Department of Medicine (Division of Experimental Medicine), Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
10
|
Li X, Zheng S, Feng Z, Liu X, Ding Y, Zhang L, Zhang G, Liu M, Zhu H, Jia H. Serum proteomics analysis of drug-naïve patients with generalised anxiety disorder: Tandem mass tags and multiple reaction monitoring. World J Biol Psychiatry 2024; 25:188-199. [PMID: 38247046 DOI: 10.1080/15622975.2023.2301064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024]
Abstract
OBJECTIVES The prevalence of generalised anxiety disorder (GAD) is high. However, the underlying mechanisms remain elusive. Proteomics techniques can be employed to assess the pathological mechanisms involved in GAD. METHODS Twenty-two drug-naive GAD patients were recruited, their serum samples were used for protein quantification and identified using Tandem Mass Tag and Multiple Reaction Monitoring (MRM). Machine learning models were employed to construct predictive models for disease occurrence by using clinical scores and target proteins as input variables. RESULTS A total of 991 proteins were differentially expressed between GAD and healthy participants. Gene Ontology analysis revealed that these proteins were significantly associated with stress response and biological regulation, suggesting a significant implication in anxiety disorders. MRM validation revealed evident disparities in 12 specific proteins. The machine learning model found a set of five proteins accurately predicting the occurrence of the disease at a rate of 87.5%, such as alpha 1B-glycoprotein, complement component 4 A, transferrin, V3-3, and defensin alpha 1. These proteins had a functional association with immune inflammation. CONCLUSIONS The development of generalised anxiety disorder might be closely linked to the immune inflammatory stress response.
Collapse
Affiliation(s)
- Xue Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Sisi Zheng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhengtian Feng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xinzi Liu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Ying Ding
- Hangzhou Seventh People's Hospital, Zhejiang, China
| | - Lina Zhang
- Hangzhou Seventh People's Hospital, Zhejiang, China
| | - Guofu Zhang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Min Liu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Hong Zhu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Hongxiao Jia
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Osorio-Conles Ó, Jiménez A, Ibarzabal A, Balibrea JM, de Hollanda A, Vidal J. Limited Bariatric Surgery-induced Weight Loss in Subjects With Type 2 Diabetes: Predictor Variables in Adipose Tissue. J Clin Endocrinol Metab 2023; 108:e1205-e1213. [PMID: 37249080 DOI: 10.1210/clinem/dgad271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Indexed: 05/31/2023]
Abstract
CONTEXT The impact of type 2 diabetes mellitus (T2D) at baseline on limited weight loss (WL) after bariatric surgery (BS) remains controversial, and the potential underlying mechanisms incompletely understood. OBJECTIVE We aimed at gaining further insight on this relationship and identifying novel associations between adipose tissue (AT) parameters and short-term WL outcomes in subjects with or without T2D undergoing BS. METHODS Mid-term WL trajectories after BS have been evaluated in a cohort of 1659 subjects (cohort 1) with (n = 543) and without T2D (n = 1116). Paired subcutaneous and visceral AT samples were obtained from a cohort of 48 pairs of subjects with and without T2D matched for age, sex, BMI, and type of BS (cohort 2). Differences in AT parameters between groups were evaluated and potential associations with WL response explored. RESULTS T2D was independently associated with a 5% lesser mid-term WL in cohort 1, while HbA1c, insulin treatment, and number of T2D medications prior to BS were only related to short-term WL outcomes. In cohort 2, a number of differentially expressed genes in AT were identified between groups, while fat cell size and fibrosis were comparable. Subcutaneous ATG7 expression was found as an independent predictor of limited WL 1 year after surgery (β: -12.21 ± 4.41, P = .008) and its addition to a clinical model significantly improved the amount of WL variability explained (R2 = 0.131 vs R2 = 0.248, F change P = .009). CONCLUSION Our results highlight the importance of T2D as determinant of limited WL following BS and suggest that dysregulated macroautophagy in subcutaneous AT may contribute to this association.
Collapse
Affiliation(s)
- Óscar Osorio-Conles
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Amanda Jiménez
- Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Ainitze Ibarzabal
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - José María Balibrea
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Ana de Hollanda
- Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Josep Vidal
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
12
|
Jääskeläinen I, Petäistö T, Mirzarazi Dahagi E, Mahmoodi M, Pihlajaniemi T, Kaartinen MT, Heljasvaara R. Collagens Regulating Adipose Tissue Formation and Functions. Biomedicines 2023; 11:biomedicines11051412. [PMID: 37239083 DOI: 10.3390/biomedicines11051412] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
The globally increasing prevalence of obesity is associated with the development of metabolic diseases such as type 2 diabetes, dyslipidemia, and fatty liver. Excess adipose tissue (AT) often leads to its malfunction and to a systemic metabolic dysfunction because, in addition to storing lipids, AT is an active endocrine system. Adipocytes are embedded in a unique extracellular matrix (ECM), which provides structural support to the cells as well as participating in the regulation of their functions, such as proliferation and differentiation. Adipocytes have a thin pericellular layer of a specialized ECM, referred to as the basement membrane (BM), which is an important functional unit that lies between cells and tissue stroma. Collagens form a major group of proteins in the ECM, and some of them, especially the BM-associated collagens, support AT functions and participate in the regulation of adipocyte differentiation. In pathological conditions such as obesity, AT often proceeds to fibrosis, characterized by the accumulation of large collagen bundles, which disturbs the natural functions of the AT. In this review, we summarize the current knowledge on the vertebrate collagens that are important for AT development and function and include basic information on some other important ECM components, principally fibronectin, of the AT. We also briefly discuss the function of AT collagens in certain metabolic diseases in which they have been shown to play central roles.
Collapse
Affiliation(s)
- Iida Jääskeläinen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Tiina Petäistö
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Elahe Mirzarazi Dahagi
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Mahdokht Mahmoodi
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Taina Pihlajaniemi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Mari T Kaartinen
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Ritva Heljasvaara
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| |
Collapse
|
13
|
Sun K, Li X, Scherer PE. Extracellular Matrix (ECM) and Fibrosis in Adipose Tissue: Overview and Perspectives. Compr Physiol 2023; 13:4387-4407. [PMID: 36715281 PMCID: PMC9957663 DOI: 10.1002/cphy.c220020] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation. It is characterized by an overaccumulation of extracellular matrix (ECM) during unhealthy expansion of adipose tissue in response to over nutrition. In obese adipose-depots, hypoxia stimulates multiple pro-fibrotic signaling pathways in different cell populations, thereby inducing the overproduction of the ECM components, including collagens, noncollagenous proteins, and additional enzymatic components of ECM synthesis. As a consequence, local fibrosis develops. The result of fibrosis-induced mechanical stress not only triggers cell necrosis and inflammation locally in adipose tissue but also leads to system-wide lipotoxicity and insulin resistance. A better understanding of the mechanisms underlying the obesity-induced fibrosis will help design therapeutic approaches to reduce or reverse the pathological changes associated with obese adipose tissue. Here, we aim to summarize the major advances in the field, which include newly identified fibrotic factors, cell populations that contribute to the fibrosis in adipose tissue, as well as novel mechanisms underlying the development of fibrosis. We further discuss the potential therapeutic strategies to target fibrosis in adipose tissue for the treatment of obesity-linked metabolic diseases and cancer. © 2023 American Physiological Society. Compr Physiol 13:4387-4407, 2023.
Collapse
Affiliation(s)
- Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Philipp E. Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
14
|
Khudyakov JI, Allen KN, Crocker DE, Trost NS, Roberts AH, Pirard L, Debier C, Piotrowski ER, Vázquez-Medina JP. Comprehensive molecular and morphological resolution of blubber stratification in a deep-diving, fasting-adapted seal. Front Physiol 2022; 13:1057721. [PMID: 36589428 PMCID: PMC9795062 DOI: 10.3389/fphys.2022.1057721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Blubber is a modified subcutaneous adipose tissue in marine mammals that provides energy storage, thermoregulation, hydrodynamic locomotion, and buoyancy. Blubber displays vertical stratification by lipid content, fatty acid composition, and vascularization, leading to the assumption that deeper blubber layers are metabolically active, while superficial layers are mainly structural and thermoregulatory. However, few studies have examined functional stratification of marine mammal blubber directly, especially in pinnipeds. We characterized morphological and transcriptional differences across blubber layers in the northern elephant seal, a deep-diving and fasting-adapted phocid. We collected blubber from seals early in their fasting period and divided blubber cores into three similarly sized portions. We hypothesized that the innermost blubber portion would have higher 1) heterogeneity in adipocyte size, 2) microvascular density, and 3) expression of genes associated with metabolism and hormone signaling than outer blubber. We found that adipocyte area and variance increased from outermost (skin-adjacent) to innermost (muscle-adjacent) blubber layers, suggesting that inner blubber has a higher capacity for lipid storage and turnover than outer blubber. Inner blubber had a higher proportion of CD144+ endothelial cells, suggesting higher microvascular density. In contrast, outer blubber had a higher proportion of CD4+ immune cells than inner blubber, suggesting higher capacity for response to tissue injury. Transcriptome analysis identified 61 genes that were differentially expressed between inner and outer blubber layers, many of which have not been studied previously in marine mammals. Based on known functions of these genes in other mammals, we suggest that inner blubber has potentially higher 1) adipogenic capacity, 2) cellular diversity, and 3) metabolic and neuroendocrine signaling activity, while outer blubber may have higher 1) extracellular matrix synthesis activity and 2) responsiveness to pathogens and cell stressors. We further characterized expression of nine genes of interest identified by transcriptomics and two adipokines with higher precision across blubber layers using targeted assays. Our study provides functional insights into stratification of blubber in marine mammals and a molecular key, including CD144, CD4, HMGCS2, GABRG2, HCAR2, and COL1A2, for distinguishing blubber layers for physiological and functional studies in seals.
Collapse
Affiliation(s)
- J. I. Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States,*Correspondence: J. I. Khudyakov,
| | - K. N. Allen
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| | - D. E. Crocker
- Department of Biology, Sonoma State University, Rohnert Park, CA, United States
| | - N. S. Trost
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - A. H. Roberts
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - L. Pirard
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - C. Debier
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - E. R. Piotrowski
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - J. P. Vázquez-Medina
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
15
|
Javed H, Singh S, Urs SUR, Oldenburg J, Biswas A. Genetic landscape in coagulation factor XIII associated defects – Advances in coagulation and beyond. Blood Rev 2022; 59:101032. [PMID: 36372609 DOI: 10.1016/j.blre.2022.101032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Coagulation factor XIII (FXIII) acts as a fine fulcrum in blood plasma that maintains the balance between bleeding and thrombosis by covalently crosslinking the pre-formed fibrin clot into an insoluble one that is resistant to premature fibrinolysis. In plasma, FXIII circulates as a pro-transglutaminase complex composed of the dimeric catalytic FXIII-A encoded by the F13A1 gene and dimeric carrier/regulatory FXIII-B subunits encoded by the F13B gene. Growing evidence accumulated over decades of exhaustive research shows that not only does FXIII play major roles in both pathological extremes of hemostasis i.e. bleeding and thrombosis, but that it is, in fact, a pleiotropic protein with physiological roles beyond coagulation. However, the current FXIII genetic-epidemiological literature is overwhelmingly derived from the bleeding pathology associated with its deficiency. In this article we review the current clinical, functional, and molecular understanding of this fascinating multifaceted protein, especially putting into the same perspective its genetic landscape.
Collapse
|
16
|
Kraposhina AY, Sobko EА, Demko IV, Kazmerchuk OV, Kacer AB, Abramov YI. The role of cathepsin S in the pathophysiology of bronchial asthma. BULLETIN OF SIBERIAN MEDICINE 2022. [DOI: 10.20538/1682-0363-2022-3-198-204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
To date, the study of the role of proteases in the pathogenesis of various diseases remains relevant. The variety of cathepsin functions is associated with the peculiarities of their localization, expression, and regulation, due to which cathepsins are involved in development of many pathologies. Dysregulation of proteases, their inhibitors, and substrates can lead to the development of multiple organ dysfunction.The review presents data on the characteristics of the entire family of cathepsins and cathepsin S, in particular. The pathophysiological role of cathepsin S in the formation of bronchopulmonary pathologies, as well as in bronchial asthma is described, and intraand extracellular implementation mechanisms are considered. The authors believe it is this enzyme that could be targeted in targeted asthma therapy to prevent airway wall remodeling at the earliest stages of the disease. The literature search was carried out in the search engines Medline, eLibrary, Scopus, the Cochrane Library, and RSCI.
Collapse
Affiliation(s)
- A. Yu. Kraposhina
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University; Krasnoyarsk Regional Clinical Hospital
| | - E. А. Sobko
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University; Krasnoyarsk Regional Clinical Hospital
| | - I. V. Demko
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University; Krasnoyarsk Regional Clinical Hospital
| | | | - A. B. Kacer
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University
| | - Yu. I. Abramov
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University
| |
Collapse
|
17
|
Michelotti TC, Kisby BR, Flores LS, Tegeler AP, Fokar M, Crasto C, Menarim BC, Loux SC, Strieder-Barboza C. Single-nuclei analysis reveals depot-specific transcriptional heterogeneity and depot-specific cell types in adipose tissue of dairy cows. Front Cell Dev Biol 2022; 10:1025240. [PMID: 36313560 PMCID: PMC9616121 DOI: 10.3389/fcell.2022.1025240] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Adipose tissue (AT) is an endocrine organ with a central role on whole-body energy metabolism and development of metabolic diseases. Single-cell and single-nuclei RNA sequencing (scRNA-seq and snRNA-seq, respectively) analyses in mice and human AT have revealed vast cell heterogeneity and functionally distinct subtypes that are potential therapeutic targets to metabolic disease. In periparturient dairy cows, AT goes through intensive remodeling and its dysfunction is associated with metabolic disease pathogenesis and decreased productive performance. The contributions of depot-specific cells and subtypes to the development of diseases in dairy cows remain to be studied. Our objective was to elucidate differences in cellular diversity of visceral (VAT) and subcutaneous (SAT) AT in dairy cows at the single-nuclei level. We collected matched SAT and VAT samples from three dairy cows and performed snRNA-seq analysis. We identified distinct cell types including four major mature adipocytes (AD) and three stem and progenitor cells (ASPC) subtypes, along with endothelial cells (EC), mesothelial cells (ME), immune cells, and pericytes and smooth muscle cells. All major cell types were present in both SAT and VAT, although a strong VAT-specificity was observed for ME, which were basically absent in SAT. One ASPC subtype was defined as adipogenic (PPARG+) while the other two had a fibro-adipogenic profile (PDGFRA+). We identified vascular and lymphatic EC subtypes, and different immune cell types and subtypes in both SAT and VAT, i.e., macrophages, monocytes, T cells, and natural killer cells. Not only did VAT show a greater proportion of immune cells, but these visceral immune cells had greater activation of pathways related to immune and inflammatory response, and complement cascade in comparison with SAT. There was a substantial contrast between depots for gene expression of complement cascade, which were greatly expressed by VAT cell subtypes compared to SAT, indicating a pro-inflammatory profile in VAT. Unprecedently, our study demonstrated cell-type and depot-specific heterogeneity in VAT and SAT of dairy cows. A better understanding of depot-specific molecular and cellular features of SAT and VAT will aid in the development of AT-targeted strategies to prevent and treat metabolic disease in dairy cows, especially during the periparturient period.
Collapse
Affiliation(s)
- Tainara C. Michelotti
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX, United States
| | - Brent R. Kisby
- Department of Pharmacology and Neuroscience, Texas Tech University Health Science Center, Lubbock, TX, United States
| | - Lauryn S. Flores
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX, United States
| | - Alexandra P. Tegeler
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX, United States
| | - Mohamed Fokar
- Center for Biotechnology and Genomics, Texas Tech University, Lubbock, TX, United States
| | - Chiquito Crasto
- Center for Biotechnology and Genomics, Texas Tech University, Lubbock, TX, United States
- Department of Computer Science, Whitacre College of Engineering, Texas Tech University, Lubbock, TX, United States
- Department of University Studies, Texas Tech University, Lubbock, TX, United States
| | - Bruno C. Menarim
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, United States
| | - Shavahn C. Loux
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, United States
| | - Clarissa Strieder-Barboza
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX, United States
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
- *Correspondence: Clarissa Strieder-Barboza,
| |
Collapse
|
18
|
Factor XIII-A in Diseases: Role Beyond Blood Coagulation. Int J Mol Sci 2021; 22:ijms22031459. [PMID: 33535700 PMCID: PMC7867190 DOI: 10.3390/ijms22031459] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 12/28/2022] Open
Abstract
Multidisciplinary research from the last few decades has revealed that Factor XIII subunit A (FXIII-A) is not only involved in blood coagulation, but may have roles in various diseases. Here, we aim to summarize data from studies involving patients with mutations in the F13A1 gene, performed in FXIII-A knock-out mice models, clinical and histological studies assessing correlations between diseases severity and FXIII-A levels, as well as from in vitro experiments. By providing a complex overview on its possible role in wound healing, chronic inflammatory bowel diseases, athe-rosclerosis, rheumatoid arthritis, chronic inflammatory lung diseases, chronic rhinosinusitis, solid tumors, hematological malignancies, and obesity, we also demonstrate how the field evolved from using FXIII-A as a marker to accept and understand its active role in inflammatory and malignant diseases.
Collapse
|